Feeds:
Posts
Comments

Archive for the ‘Nitric Oxide in Health and Disease’ Category

Metabolic Genomics and Pharmaceutics, Vol. 1 of BioMed Series D available on Amazon Kindle

Metabolic Genomics and Pharmaceutics, Vol. 1 of BioMed Series D available on Amazon Kindle

Reporter: Stephen S Williams, PhD

Article ID #180: Metabolic Genomics and Pharmaceutics, Vol. 1 of BioMed Series D available on Amazon Kindle. Published on 8/15/2015

WordCloud Image Produced by Adam Tubman

Leaders in Pharmaceutical Business Intelligence would like to announce the First volume of their BioMedical E-Book Series D:

Metabolic Genomics & Pharmaceutics, Vol. I

SACHS FLYER 2014 Metabolomics SeriesDindividualred-page2

which is now available on Amazon Kindle at

http://www.amazon.com/dp/B012BB0ZF0.

This e-Book is a comprehensive review of recent Original Research on  METABOLOMICS and related opportunities for Targeted Therapy written by Experts, Authors, Writers. This is the first volume of the Series D: e-Books on BioMedicine – Metabolomics, Immunology, Infectious Diseases.  It is written for comprehension at the third year medical student level, or as a reference for licensing board exams, but it is also written for the education of a first time baccalaureate degree reader in the biological sciences.  Hopefully, it can be read with great interest by the undergraduate student who is undecided in the choice of a career. The results of Original Research are gaining value added for the e-Reader by the Methodology of Curation. The e-Book’s articles have been published on the Open Access Online Scientific Journal, since April 2012.  All new articles on this subject, will continue to be incorporated, as published with periodical updates.

We invite e-Readers to write an Article Reviews on Amazon for this e-Book on Amazon.

All forthcoming BioMed e-Book Titles can be viewed at:

http://pharmaceuticalintelligence.com/biomed-e-books/

Leaders in Pharmaceutical Business Intelligence, launched in April 2012 an Open Access Online Scientific Journal is a scientific, medical and business multi expert authoring environment in several domains of  life sciences, pharmaceutical, healthcare & medicine industries. The venture operates as an online scientific intellectual exchange at their website http://pharmaceuticalintelligence.com and for curation and reporting on frontiers in biomedical, biological sciences, healthcare economics, pharmacology, pharmaceuticals & medicine. In addition the venture publishes a Medical E-book Series available on Amazon’s Kindle platform.

Analyzing and sharing the vast and rapidly expanding volume of scientific knowledge has never been so crucial to innovation in the medical field. WE are addressing need of overcoming this scientific information overload by:

  • delivering curation and summary interpretations of latest findings and innovations on an open-access, Web 2.0 platform with future goals of providing primarily concept-driven search in the near future
  • providing a social platform for scientists and clinicians to enter into discussion using social media
  • compiling recent discoveries and issues in yearly-updated Medical E-book Series on Amazon’s mobile Kindle platform

This curation offers better organization and visibility to the critical information useful for the next innovations in academic, clinical, and industrial research by providing these hybrid networks.

Table of Contents for Metabolic Genomics & Pharmaceutics, Vol. I

Chapter 1: Metabolic Pathways

Chapter 2: Lipid Metabolism

Chapter 3: Cell Signaling

Chapter 4: Protein Synthesis and Degradation

Chapter 5: Sub-cellular Structure

Chapter 6: Proteomics

Chapter 7: Metabolomics

Chapter 8:  Impairments in Pathological States: Endocrine Disorders; Stress

                   Hypermetabolism and Cancer

Chapter 9: Genomic Expression in Health and Disease 

 

Summary 

Epilogue

 

 

Read Full Post »

Angiogenesis Inhibitors [9.5]

Writer and Curator: Larry H Bernstein, MD, FCAP

This article has the following structure:

9.5.1 Motesanib (AMG 706)

9.5.2 Drugs that block cancer blood vessel growth (anti angiogenics)

9.5.3 Recent Advances in Anti-Angiogenic Therapy of Cancer

9.5.4 Angiogenesis inhibitors in cancer therapy: mechanistic perspective on classification and treatment rationales

9.5.5 LUCITANIB a VEGFR/FGFR dual kinase inhibitor in Phase 2 trials

9.5.1 Motesanib (AMG 706)

by DR ANTHONY MELVIN CRASTO Ph.D

http://newdrugapprovals.org/2015/05/15/motesanib-amg-706/

Motesanib (AMG 706) is an experimental drug candidate originally developed by Amgen[1] but is now being investigated by theTakeda Pharmaceutical Company. It is an orally administered small molecule belonging to angiokinase inhibitor class which acts as an antagonist of VEGF receptorsplatelet-derived growth factor receptors, and stem cell factor receptors.[2] It is used as the phosphate salt motesanib diphosphate.

Motesanib, also known as AMG-706, is an orally administered multikinase inhibitor that selectively targets VEGF receptors, platelet-derived growth factor receptors, and Kit receptors.

N-(3,3-Dimethylindolin-6-yl){2-[(4-pyridylmethyl)amino](3-pyridyl)}carboxamide

motesanib-amg-706-a10608

motesanib-amg-706-a10608

http://www.adooq.com/media/catalog/product/cache/1/image/9df78eab33525d08d6e5fb8d27136e95/m/o/motesanib-amg-706-a10608.gif

http://www.chemblink.com/products/453562-69-1.htm

9.5.2 Drugs that block cancer blood vessel growth (anti angiogenics)

http://www.cancerresearchuk.org/about-cancer/cancers-in-general/treatment/biological/types/drugs-that-block-cancer-blood-vessel-growth

When it has reached 1 to 2mm across, a tumor needs to grow its own blood vessels in order to continue to get bigger. Some cancer cells make a protein called vascular endothelial growth factor (VEGF). The VEGF protein attaches to receptors on cells that line the walls of blood vessels within the tumour.

Drugs that block blood vessel growth factor

Some drugs block vascular endothelial growth factor (VEGF) from attaching to the receptors on the cells that line the blood vessels. This stops the blood vessels from growing.

A drug that blocks VEGF is bevacizumab (Avastin). It is also a monoclonal antibody.

Drugs that block signalling within the cell

Some drugs stop the VEGF receptors from sending growth signals into the blood vessel cells. These treatments are also called cancer growth blockers or tyrosine kinase inhibitors (TKIs).

Sunitinib (Sutent) is a type of TKI that blocks the growth signals inside blood vessel cells. It is used to treat kidney cancer and a rare type of stomach cancer called gastrointestinal stromal tumour (GIST).

Drugs that affect signals between cells

Some drugs act on the chemicals that cells use to signal to each other to grow. This can block the formation of blood vessels. Drugs that works in this way include thalidomide and lenalidomide (Revlimid).

Each drug has different side effects. You can look up the name of your drug in our cancer drug section to find out about the side effects you may have.

To find trials using anti angiogenesis treatment go to our clinical trials database and type ‘angiogenesis’ into the search box.

http://www.cancer.gov/about-cancer/treatment/types/immunotherapy/angiogenesis-inhibitors-fact-sheet

Tumors can cause their blood supply to form by giving off chemical signals that stimulate angiogenesis. Tumors can also stimulate nearby normal cells to produce angiogenesis signaling molecules. The resulting new blood vessels “feed” growing tumors with oxygen and nutrients, allowing the cancer cells to invade nearby tissue, to move throughout the body, and to form colonies of cancer cells, called metastases. Because tumors cannot grow beyond a certain size or spread without a blood supply, scientists are trying to find ways to block tumor angiogenesis.

Angiogenesis requires the binding of signaling molecules, such as vascular endothelial growth factor (VEGF), to receptors on the surface of normal endothelial cells. When VEGF and other endothelial growth factors bind to their receptors on endothelial cells, signals within these cells are initiated that promote the growth and survival of new blood vessels.

Angiogenesis inhibitors interfere with various steps in this process. For example, bevacizumab (Avastin®) is a monoclonal antibody that specifically recognizes and binds to VEGF (1). When VEGF is attached to bevacizumab, it is unable to activate the VEGF receptor. Other angiogenesis inhibitors, including sorafenib and sunitinib, bind to receptors on the surface of endothelial cells or to other proteins in the downstream signaling pathways, blocking their activities (2).

The U.S. Food and Drug Administration (FDA) has approved bevacizumab to be used alone forglioblastoma that has not improved with other treatments and to be used in combination with other drugs to treat metastatic colorectal cancer, some non-small cell lung cancers, and metastatic renal cell cancer. Bevacizumab was the first angiogenesis inhibitor that was shown to slow tumor growth and, more important, to extend the lives of patients with some cancers.

The FDA has approved other drugs that have antiangiogenic activity, including sorafenib (Nexavar®), sunitinib(Sutent®), pazopanib (Votrient®), and everolimus (Afinitor®). Sorafenib is approved for hepatocellular carcinoma and kidney cancer, sunitinib and everolimus for both kidney cancer and neuroendocrine tumors, and pazopanib for kidney cancer.

Angiogenesis inhibitors are unique cancer-fighting agents because they tend to inhibit the growth of blood vessels rather than tumor cells. In some cancers, angiogenesis inhibitors are most effective when combined with additional therapies, especially chemotherapy. It has been hypothesized that these drugs help normalize the blood vessels that supply the tumor, facilitating the delivery of other anticancer agents, but this possibility is still being investigated.

Angiogenesis inhibitor therapy does not necessarily kill tumors but instead may prevent tumors from growing. Therefore, this type of therapy may need to be administered over a long period.

Initially, it was thought that angiogenesis inhibitors would have mild side effects, but more recent studies have revealed the potential for complications that reflect the importance of angiogenesis in many normal body processes, such as wound healing, heart and kidney function, fetal development, and reproduction. Side effects of treatment with angiogenesis inhibitors can include problems with bleeding, clots in the arteries (with resultant stroke or heart attack), hypertension, and protein in the urine (35). Gastrointestinal perforation and fistulas also appear to be rare side effects of some angiogenesis inhibitors.

In addition to the angiogenesis inhibitors that have already been approved by the FDA, others that target VEGF or other angiogenesis pathways are currently being tested in clinical trials (research studies involving patients). If these angiogenesis inhibitors prove to be both safe and effective in treating human cancer, they may be approved by the FDA and made available for widespread use.

In addition, phase I and II clinical trials are testing the possibility of combining angiogenesis inhibitor therapy with other treatments that target blood vessels, such as tumor-vascular disrupting agents, which damage existing tumor blood vessels (6).

9.5.3 Recent Advances in Anti-Angiogenic Therapy of Cancer

Rajeev S. Samant and Lalita A. Shevde
Oncotarget. 2011 Mar; 2(3): 122–134.
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3260813/

More than forty anti-angiogenic drugs are being tested in clinical trials all over the world. This review discusses agents that have approved by the FDA and are currently in use for treating patients either as single-agents or in combination with other chemotherapeutic agents.

Tumor angiogenesis is generation of a network of blood vessels within the cancerous growth. This process can occur two ways: The more accepted model involves the release of signaling molecules by the tumor cells; these molecules activate the surrounding tissue to promote growth of new blood vessels. This stimulates vascular endothelial cells to divide rapidly [910]. The other model proposes the generation of new vasculature by vasculogenic mimicry. This model argues that the tumor cells trans-differentiate in endothelial-like cells and create structures from inside of the tumor tapping into a nearby blood vessel [4].

Escape of the tumor cell from the confines of the primary tumor to distant body parts is the pre-requisite for hematogenous metastasis. This escape route is provided by the tumor vasculature. Thus, it was envisioned that inhibition of angiogenesis will also lead to inhibition of metastasis. This phenomenon was demonstrated by very elegant mouse model studies using angiostatin [1112]. Angiostatin was also demonstrated to be secreted by some primary tumors leading to restricted growth of the metastasis leading to “dormancy” of the metastasis. Mice deficient in angiogenesis (Id1 & Id3 deficient) showed significantly less tumor take rates [13]. Independent studies showed absence of metastasis in angiogenesis deficient mice [1415]. Defective angiogenesis was attributed to impaired VEGF-dependent recruitment of precursor endothelial cells from the bone marrow to the newly developing tumor vasculature [16].

Metastasis of malignant tumors to regional lymph nodes is one of the early signs of cancer spread in patients, and it occurs at least as frequently as hematogenous metastasis [17]. Particularly, in cancers, such as breast cancer, lymphatic metastasis is a predominant route for tumor spread. The contribution of lymphatic system to the tumor growth is an area that is relatively less studied. However, lymphatic vessels are speculated to contribute to tumor growth and metastasis in a variety of ways. The VEGF, FGF2 and PDGF produced by vascular endothelial cells are proposed to be involved in the activation of lymphatic endothelial cells, which in turn produce matrix metalloproteases and urokinase plasminogen activator (uPA) that can promote malignant tumor growth. Thus, there exists a synergistic crosstalk between the tumor and the lymphatic vessels and blood vessels.

Angiogenesis is a complex and intricately regulated process. Like all other regulated biological phenomena, angiogenesis has activators or pro-angiogenic factors and inhibitors or anti-angiogenic factors [9].

The Activators

Tumor cells activate signaling pathways that promote uncontrolled proliferation and survival. These include the PI3K/AKT/mTOR pathway, Hedgehog pathway and, Wnt pathway [1824] that produce pro-angiogenic signaling intermediates [2526]. Among the several reported activators of angiogenesis present in cells two proteins appear to be the most important for sustaining tumor growth: vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF). VEGF and bFGF are secreted by the tumor into the surrounding tissue. They bind to their cognate receptors on endothelial cells. This activates a signaling cascade that transmits a nuclear signal prompting target genes to activate endothelial cell growth. Activated endothelial cells also produce matrix metalloproteinases (MMPs). These MMPs break down the extracellular matrix and allow the migration of endothelial cells. The division and migration of the endothelial cells leads to formation of new blood vessels [2728].

The Inhibitors

If angiogenesis is so critical for the tumor growth, then agents that inhibit angiogenesis would have great therapeutic value. With the discovery of endostatin, the concept of anti-angiogenic therapy was launched and popularized by Dr. Folkman [29]. Angiogenesis inhibitors have been discovered from a variety of sources. Some are naturally present in the human body e.g. specific fragments of structural proteins such as collagen or plasminogen (angiostatin, endostatin, tumstatin) [30]. Others are natural products in green tea, soy beans, fungi, mushrooms, tree bark, shark tissues, snake venom etc. [31]. A plethora of synthetic compounds are also characterized to have anti-angiogenic properties [32].

ANTI-ANGIOGENIC TREATMENT OF CANCER

Since angiogenesis is an event critical to primary tumor growth as well as metastasis, anti-angiogenic treatment of tumors is a highly promising therapeutic avenue [33]. Thus, for over last couple of decades, there has been a robust activity aimed towards the discovery of angiogenesis inhibitors [3435]. More than forty anti-angiogenic drugs are being tested in human cancer patients in clinical trials all over the world. From the several anti-angiogenic agents reported, we have focused this review on discussing those agents that have received FDA approval in the United States and are currently in use for treating patients either as a single-agent or in combination with other chemotherapeutic agents (Figure ​(Figure1).1). Based on functionality, the anti-angiogenic drugs can be sub-divided into three main groups:

angiogenesis inhibitors oncotarget-02-122-g001

angiogenesis inhibitors oncotarget-02-122-g001

Figure 1

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3260813/bin/oncotarget-02-122-g001.jpg

Targets of FDA-approved angiogenesis inhibitors: Angiogenesis inhibitors impact both, the tumor as well as the endothelial cells resulting in the disruption of the effects of the microenvironment in promoting tumor growth and angiogenesis

Drugs that inhibit growth of endothelial cells

e.g. Endostatin and combretastatin A4, cause apoptosis of the endothelial cells [36]. Thalidomide is also a potent inhibitor of endothelial cell growth [37].

Drugs that block angiogenesis signaling

e.g. anti-VEGF antibodies (Avastin, FDA approved for colorectal cancer), Interferon-alpha (inhibits the production of bFGF and VEGF) [36].

Drugs that block extracellular matrix breakdown

e.g. inhibitors of MMPs [38].

ANTI-ANGIOGENIC THERAPIES THAT HAVE RECEIVED USA-FDA APPROVAL

Conventional chemotherapy is usually a systemic therapy that tries to capture a narrow therapeutic window offered by rapid proliferation of tumor cells compared to the normal cells. Chemotherapy has significant side effects such as hair loss, diarrhea, mouth ulcer, infection, and low blood counts. Anti-angiogenic therapy has several advantages over chemotherapy as it is mostly not directed towards directly killing cells but stopping the blood vessel formation, an event that is rare in tissues other than growing tumor. Hence it is well tolerated by the patients and has fewer side effects [29]. There are currently seven approved anti-cancer therapies in two primary categories:

  1. Monoclonal antibodies directed against specific pro-angiogenic growth factors and/or their receptors
  2. Small molecule tyrosine kinase inhibitors (TKIs) of multiple pro-angiogenic growth factor receptors.

Besides these, inhibitors of mTOR (mammalian target of rapamycin), proteasome inhibitors and thalidomide have also been reported to indirectly inhibit angiogenesis through mechanisms that are not completely understood.

MONOCLONAL ANTIBODY THERAPIES

Four monoclonal antibody therapies are approved to treat several tumor types:

Bevacizumab (Avastin®)

The first FDA approved angiogenesis inhibitor, Avastin is a humanized monoclonal antibody that binds biologically active forms of vascular endothelial growth factor (VEGF) and prevents its interaction with VEGF receptors (VEGFR-1 and VEGFR-2), thereby inhibiting endothelial cell proliferation and angiogenesis. Bevacizumab has been tested in phase I studies in combination with chemotherapy with a good safety profile [39]. This treatment is approved for metastatic colorectal cancer and non-small cell lung cancer [4043]. Bevacizumab has also evolved as a first line of treatment in combination with paclitaxel in breast cancer patients by virtue of its ability to double median progression-free survival (PFS) [44]. In combination with chemoendocrine therapy (including capecitabine and vinorelbine, and letrozole) bevacizumab treatment significantly decreased the percentage of viable circulating endothelial cells and prevented the chemotherapy-induced mobilization of circulating progenitors [45]. In combination with irinotecan, bevacizumab significantly increased PFS in glioma patients [4647]. VEGF has emerged as a compelling therapeutic target for leukemias. Inhibition of angiogenesis in hematological malignancies interdicts the angiogenesis within the bone marrow ecosystem comprised of multiple cell types, including fibroblasts, endothelial progenitor cells, endothelial cells, dendritic cells and, malignant cells, blocking the availability of nutrients to cancer cells and disrupting crosstalk between the various cell types to curtail the malignant phenotype [48].

Cetuximab (Erbitux®)

This is a monoclonal antibody that binds the extracellular domain of epidermal growth factor receptor (EGFR), preventing ligand binding and activation of the receptor resulting in internalization and degradation of the receptor culminating in inhibition of cell proliferation and angiogenesis. Cetuximab downregulated VEGF expression in a dose-dependent manner in a human colorectal carcinoma (CRC) cell line and in human CRC mouse xenografts [49]. The xenografts also showed a significant reduction in blood vessel counts following several rounds of cetuximab treatment [49], indicating that the tumor-promoting effects of EGFR overexpression may be mediated through VEGF stimulation and tumor angiogenesis. This treatment is approved for metastatic CRC and head and neck cancer [50] in patients who are refractory to irinotecan-based chemotherapy. In combination with irinotecan (an inhibitor of topoisomerase I), cetuximab is the first monoclonal antibody that has been approved by the FDA as second-line treatment for metastatic colorectal cancer [5152]. In Phase I and Phase III trials [5354] cetuximab significantly improved the effects of radiotherapy in patients with unresectable (cannot be removed by surgery) squamous cell carcinoma of the head and neck (SCCHN). Cetuximab has also been shown to sensitize cells to radiation and chemotherapy, potentially through blocking EGFR nuclear import and the associated activation of DNA protein kinase enzymes necessary for repairing radiation- and chemotherapy-induced DNA damage [55]. Compared to radiation alone, cetuximab plus radiation therapy can nearly double the median survival in patients with a certain kind of head and neck cancer that has not spread to other parts of the body [54] making cetuximab the only drug achieving interesting response rate in second line treatment of advanced SCCHN [56]. Cetuximab was also found to be tolerated well in combination with cisplatin, or carboplatin, and fluorouracil [5758].

Panitumumab (Vectibix™)

It is a fully humanized anti-EGFR monoclonal antibody that binds specifically to the human EGFR. Panitumumab is a recombinant human monoclonal antibody [59]; therefore, the risk of an infusion reaction is minimized. Vectibix® is indicated as a single agent for the treatment of EGFR-expressing, metastatic colorectal carcinoma with disease progression on or following fluoropyrimidine-, oxaliplatin-, and irinotecan-containing chemotherapy regimens [6062]. The effectiveness of Vectibix® as a single agent for the treatment of EGFR-expressing, metastatic CRC is based on progression-free survival [6364]. Panitumumab is used in patients who are not responding to regimens containing fluorouracil, oxaliplatin, and irinotecan [60]. Patients often receive panitumumab after receiving bevacizumab or cetuximab. Panitumumab can be given with FOLFOX (oxaliplatin, leucovorin, and fluorouracil) or FOLFIRI (irinotecan, leucovorin, and fluorouracil) regimens, or as a single agent. Currently no data are available that demonstrate an improvement in disease-related symptoms or increased survival with Vectibix® in colon cancer [65]. This drug is also being tested for aerodigestive track and head and neck cancer [6667].

Trastuzumab (Herceptin®)

Is a humanized monoclonal antibody that binds the extracellular domain of HER-2, which is overexpressed in 25-30% of invasive breast cancer tumors [68]. HER2-positive breast cancer is highly aggressive disease with high recurrence rate, poorer prognosis with decreased survival compared with HER2-negative breast cancer [69]. Herceptin® is designed to target and block the function of HER2 protein overexpression. This is the first humanized antibody is approved for Breast cancer [70]. Herceptin® is approved by the FDA to treat HER2 positive breast cancer that has metastasized after treatment with other anticancer drugs [71]. It is also approved to be used with other drugs to treat HER2-positive breast cancer that has spread to the lymph nodes to be used after surgery. The FDA first approved Herceptin in September 1998 [7173]. In November 2006, the FDA approved Herceptin as part of a treatment regimen containing doxorubicin, cyclophosphamide and paclitaxel, for the adjuvant treatment of patients with HER2-positive, node-positive breast cancer (http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/default.htm). In January 2008, the FDA approved Herceptin as a single agent for the adjuvant treatment of HER2-overexpressing node-negative (ER/PR-negative or with one high-risk feature) or node-positive breast cancer, following multi-modality anthracycline-based therapy (http://biopharminternational.findpharma.com/biopharm/News/FDA-Approves-Expanded-Adjuvant-Indications-for-Her/ArticleStandard/Article/detail/518867). Trastuzumab is also being studied in the treatment of other types of cancers such as pancreatic [74], endometrial [75], lung [76], cervical [77] and ovarian cancer [78]

SMALL MOLECULE TYROSINE KINASE INHIBITORS (TKIs)

Protein tyrosine kinases have emerged as crucial targets for therapeutic intervention in cancer especially because they play an important role in the modulation of growth factor signaling. As per ClinicalTrials.gov (www.clinicaltrials.gov), there are 43 ongoing studies on tyrosine kinase inhibitors in angiogenesis. Since discussing all of them is beyond the scope of this article, we have focused our discussion on the three TKIs that are currently approved as anti-cancer therapies:

Erlotinib (Tarceva®)

Erlotinib hydrochloride (originally coded as OSI-774) is an orally available, potent, reversible, and selective inhibitor of the EGFR (ErbB1) tyrosine kinase activity. Erlotinib hydrochloride has been approved by FDA for treatment of patients with locally advanced or metastatic NSCLC after failure of at least one prior chemotherapy regimen [7980]. Interesting recent studies have demonstrated that since Erlotinib and Bevacizumab act on two different pathways critical to tumor growth and dissemination, administering these drugs concomitantly may confer additional clinical benefits to cancer patients with advanced disease. This combination therapy may prove to be a viable second-line alternative to chemotherapy in patients with NSCLC [81]. Also, for patients with locally advanced, unresectable or metastatic pancreatic carcinoma, Erlotinib has received FDA approval for the treatment in combination with gemcitabine [8283]. Erlotinib is also being studied in the treatment of other types of cancers. For example combination of Erlotinib with Bevacizumab has been evaluated in metastatic breast cancer [84], hepatocellular carcinoma [85] and in metastatic renal cancer [86] as phase II trials. Outcomes for prostate, cervical and colorectal cancers treated with Erlotinib are cautiously optimistic [8789].

Sorafenib (Nexavar®)

Sorafenib is an orally active inhibitor of VEGFR-1, VEGFR-2, VEGFR-3, PDGFR-β, and Raf-1 tyrosine kinase activity [90]. It has received the approval of FDA for the treatment of patients with unresectable hepatocellular carcinoma [91] and advanced renal cell carcinoma [92]. However, not all advanced hepatocellular carcinoma patients were able to tolerate sorafenib and some patients experienced tumor progression [91]. Sorafenib has shown improvements in PFS in patients with renal cell carcinoma [93]. It is one of the aggressively studied drugs. According to the NCI clinical trials search results, there are about 168 active clinical trials involving sorafenib in a variety of cancers.

Sunitinib (Sutent®)

Sunitinib targets activity of multiple tyrosine kinases such as VEGFR-1, VEGFR-2, VEGFR-3, PDGFR- β, and RET [94]. It is approved by FDA as Sunitinib malate for treating advanced (metastatic) renal cell carcinoma [95]. It is also approved by FDA for gastrointestinal stromal tumor (GIST) in patients whose disease has progressed or who are unable to tolerate treatment with imatinib (Gleevec), the current treatment for GIST patients [9596]. Sunitinib has shown early evidence of anti-tumor activity in Phase II trials in US, European and Asian patients with locally advanced, unresectable and metastatic hepatocellular carcinoma. A Phase III trial of sunitinib in hepatocellular carcinoma is ongoing [97]. According to the NCI clinical trials search results, Sunitinib is currently evaluated in about 150 active clinical trials. It is evaluated for ovarian [98], breast [99] and non small cell lung cancer [100] among others [101].

Inhibitors of mTOR

mTOR plays a part in the PI3 kinase/AKT pathway involved in tumor cell proliferation and angiogenesis [102]. Rapamycin and related mTOR inhibitors inhibit endothelial cell VEGF expression, as well as VEGF-induced endothelial cell proliferation [103]. Inhibitors of mTOR are an important class of anti-angiogenic agents. These include: deforolimus, everolimus, rapamycin (sirolimus), and temsirolimus [104105]. Temsirolimus (Toricel™) is a small molecule inhibitor of mTOR, approved for treating advanced renal cell carcinoma [106]. It is a type of rapamycin analog and a type of serine/threonine kinase inhibitor, it is also called CCI-779. In pre-clinical models combination therapy for treating breast cancer using anti-estrogen, ERA-923, and temsirolimus has been successfully tested [107]. It is found to be highly effective against human melanoma when tested in combination with cisplatin and DTIC (in independent studies) in a SCID mouse xenotranplantation model [108109]. There are over 41 active studies of Temsirolimus for a variety of solid tumors [110]. mTOR inhibition has also been strongly advocated in as a putative cancer therapeutic strategy for urologic malignancies [111]. In a pilot study (6 patients) with imatinib-resistant CML, rapamycin induced major and minor leukocyte responses, with an observed decrease in the mRNA levels of VEGFA in circulating leukaemic cells [112]. Combination treatments for breast cancer with aromatase inhibitor [113] and letrozol [114] are also being evaluated. Rapamycin treatment brought partial responses (>50% reduction in the absolute number of blood blasts) and stable disease in adult refractory/relapsed AML [115]. In a recent report, Deforolimus was studied in a Phase 2 trial in pretreated patients with various hematological malignancies, including ALL, AML, CLL, CML, MDS, agnogenic myeloid metaplasia, mantle cell lymphoma and T-cell leukemia/lymphoma [116]. Overall, 40% of deforolimus-treated patients experienced hematological improvement or stable disease.

OTHER ANGIOGENIC AGENTS

Bortezomib (Velcade®)

Is a proteasome inhibitor that disrupts signaling of cancer cells, leading to cell death and tumor regression. It is the first compound in its class to be used in clinical practice. It has indirect anti-angiogenic properties [117]. While its exact mechanism is not understood, it induces the pro-apoptotic BH3-only family member NOXA in a p53 independent fashion triggering of a caspase cascade culminating in apoptosis in melanoma and myeloma cells [118]. It is FDA-approved for the treatment of myeloma that has relapsed after two prior treatments (or where resistance has developed following the last treatment). It was also found to induce high quality responses as third line salvage therapy with acceptable toxicity in a significant proportion of homogeneously pre-treated myeloma patients with progressive disease after autologous transplantation and thalidomide. [119]. In a Phase 3 trial involving 669 myeloma patients treated with at least one prior therapy, bortezomib increased median, improved overall survival, and increased response rate, compared with high-dose dexamethasone [120]. In combination with doxorubicin and gemcitabine, bortezomib was also found to be effective in heavily pretreated, advanced Cutaneous T cell Lymphomas (CTCL) [121]. Bortezomib was also reported to be active as a single agent for patients with relapsed/refractory CTCL and Peripheral T Cell Lymphoma (PTCL) with skin involvement [122]. On the contrary, the use of bortezomib was discouraged after a phase II study revealed that found in combination with dexamethasone, bortezomib is not active in heavily pre-treated patients with relapsed Hodgkin’s lymphoma [123124].

Thalidomide (Thalomid®)

Possesses immunomodulatory, anti-inflammatory, and anti-angiogenic properties, although the precise mechanisms of action are not fully understood. Thalidomide was the first angiogenesis inhibitor to demonstrate clinical efficacy in multiple myeloma [37125]. Specifically in myeloma, thalidomide down-regulated VEGF secretion from bone marrow endothelial cells obtained from patients with active disease. In a landmark Phase 2 clinical trial, 169 previously treated patients with refractory myeloma received thalidomide monotherapy [126]. Partial response, was achieved in 30% of patients, and 14% achieved a complete or nearly complete remission. The survival rate at 2 years was 48%. These results led to many subsequent clinical studies of thalidomide in myeloma, leading ultimately to FDA approval of the drug in 2006, for the treatment of newly diagnosed multiple myeloma, in combination with dexamethasone. In the pivotal Phase 3 trial, the response rate in patients receiving thalidomide plus dexamethasone was 63% compared to 41% with dexamethasone alone [127]. Long-term outcome measures, including time-to-progression (TTP) and PFS, were recently reported for a 470 patient randomized, placebo-controlled Phase 3 clinical trial of a similar protocol in newly diagnosed multiple myeloma, with comparable overall response rates [128]. Significant increases resulted in both median TTP and median PFS for the thalidomide plus dexamethasone group versus dexamethasone alone.

Thalidomide was found to be moderately tolerated and minimally effective in patients with histologically proven advanced hepatocellular carcinoma [129]. Thalidomide provided no survival benefit for patients with multiple, large, or midbrain metastases when combined with WBRT (whole-brain radiation therapy) [130]. On the contrary, thalidomide did not significantly add to the efficacy of the fludarabine, carboplatin, and topotecan (FCT) regimen in poor prognosis AML patients [131] and was also ineffective in improving prognosis or decreasing plasma VEGF levels in patients with persistent or recurrent leiomyosarcoma of the uterus [132].

METRONOMIC THERAPY

While conventional anti-angiogenic therapy is based on Maximum Tolerated Doses (MTD), the cells involved in angiogenesis may regenerate during the three- to four-week interval between cycles of the chemotherapy. Taking advantage of the fact that endothelial cells are about 10–100 times more susceptible to chemotherapeutic agents than cancer cells, therapy based on daily, oral, low-dose chemotherapeutic drugs was designed. Metronomic chemotherapy refers to the close, rhythmic administration of low doses of cytotoxic drugs, with minimal or no drug-free breaks, over prolonged periods. Metronomic therapy appears promising mainly due to the fact that its anti-angiogenic and anti-tumorigenic effects are accompanied by low toxicity, limited side effects, no need for hospitalization and allowing for feasible combinations with selective inhibitors of angiogenesis. There are several foreseeable advantages and opportunities for metronomic chemotherapy: activity against the parenchymal and stromal components, pro-apoptotic activity, reduction of the likelihood of emergence of acquired resistance, feasibility of long term administration and acceptable systemic side effects [133]. In a pilot phase II study conducted by Correale et al [134] to investigate the toxicity and activity of the novel metronomic regimen of weekly cisplatin and oral etoposide in high-risk patients with NSCLC, the objective response rate was 45.2%, disease control was 58.1%, meantime to progression and survival were 9 and 13 months, respectively. Pharmacokinetic analysis showed that this regimen allowed a greater median monthly area under the curve of the drugs than conventional schedules. In a Phase I trial of metronomic dosing of docetaxel and thalidomide, of the 26 patients with advanced tumors enrolled, prolonged freedom from disease progression was observed in 44.4% of the evaluable patients [135].

Circulating endothelial progenitor cells (EPCs) also participate in tumor angiogenesis. In a study comparing the effects of metronomic chemotherapy over conventional dose-dense chemotherapy, it was found that the numbers of circulating EPCs and the plasma levels of VEGF increased sharply, doubling pre-therapeutic levels at day 21 after conventional chemotherapy, whereas under low-dose metronomic chemotherapy, the numbers of circulating EPCs decreased significantly and VEGF plasma concentrations remained unchanged. These observations provide evidence that conventional dose-dense chemotherapy leads to rebound EPC mobilization even when given with adjuvant intention, while low-dose metronomic scheduling of cytotoxic substances such as trofosfamide may sharply reduce EPC release into the circulation. [136].

Combined bevacizumab and metronomic oral cyclophosphamide was also discovered to be a safe and effective regimen for heavily pre-treated ovarian cancer patients [137]. Treatment with metronomic capecitabine and cyclophosphamide in combination with bevacizumab was shown to be effective in advanced breast cancer and additionally was minimally toxic [138]. Metronomic treatment with carboplatin and vincristine associated with fluvastatin and thalidomide significantly increased survival of pediatric brain stem tumor patients. Tumor volume showed a significant reduction accompanied by increased quality of life [139]. Thus, given the fact that the most evident effect of selective anti-angiogenic agents (i.e. bevacizumab) is the significant prolonging of the duration of response obtainable by chemotherapy alone, with minimal possible side effects of cytotoxic agents given in association metronomic chemotherapy should be considered both as novel up-front or maintenance treatment in patients with biologically poorly aggressive advanced cancer diseases [140].

Overall, metronomic chemotherapy was able to induce tumor stabilization and prolong the duration of clinical benefit, without much associated toxicity. Emerging evidence suggests that metronomic chemotherapy could also activate the host immune system and potentially induce tumor dormancy [141143].

CONCLUSIONS AND FUTURE PERSPECTIVES

While angiogenesis as a hallmark of tumor development and metastasis is now a validated target for cancer treatment, the overall benefits of anti-angiogenic drugs from the perspective of impacting survival have left much to desire, endorsing a need for developing more effective therapeutic regimens e.g., combining anti-angiogenic drugs with established chemotherapeutic drugs [144145]. There are now several agents that target the tumor vasculature through different pathways, either by inhibiting formation of the tumor neovasculature or by directly targeting the mature tumor vessels. The main body of evolving evidence suggests that their effects are compounded by their synergistic use with conventional chemotherapy rather than individual agents. Anti-angiogenic drugs such as bevacizumab can bring about a transient functional normalization of the tumor vasculature. This can have an additive effect when co-administered with chemo/radiotherapy. But long term inhibition of angiogenesis reduces tumor uptake of co-administered chemotherapeutic agents. This underscores the need for discovering new targets for anti-angiogenic therapy in order to effectively prohibit angiogenesis and circumvent mechanisms that contribute to resistance mechanisms that emerge with long term use of anti-angiogenic therapies. It also warrants a need to define reliable surrogate indicators of effectiveness of the anti-angiogenic therapy as well as dependable markers for identifying the patients who are most likely to benefit from the combination of anti-angiogenic therapy and conventional chemotherapy.

Several new frontiers are emerging. New advances in understanding endothelial cells, which constitute the tumor vasculature, towards developing antiangiogenic strategies are one of the important ones [146147]. Novel cellular targets such as integrins and microRNAs and novel treatment options such as possible use of pharmaconutrients to modulate angiogenic pathways need careful testing and evaluation [148151]. Finally, the administration of these drugs in a metronomic schedule is likely to improve the overall response to anti-angiogenic drugs making it feasible to administer them with conventionally toxic chemotherapeutic drugs, thus increasing the armamentarium of drug combinations that can be employed for treatment.

9.5.4 Angiogenesis inhibitors in cancer therapy: mechanistic perspective on classification and treatment rationales

El-Kenawi AE1, El-Remessy AB.
Br J Pharmacol. 2013 Oct; 170(4):712-29.
http://dx.doi.org:/10.1111/bph.12344

Angiogenesis, a process of new blood vessel formation, is a prerequisite for tumor growth to supply the proliferating tumor with oxygen and nutrients. The angiogenic process may contribute to tumour progression, invasion and metastasis, and is generally accepted as an indicator of tumor prognosis. Therefore, targeting tumor angiogenesis has become of high clinical relevance. The current review aimed to highlight mechanistic details of anti-angiogenic therapies and how they relate to classification and treatment rationales. Angiogenesis inhibitors are classified into either direct inhibitors that target endothelial cells in the growing vasculature or indirect inhibitors that prevent the expression or block the activity of angiogenesis inducers. The latter class extends to include targeted therapy against oncogenes, conventional chemotherapeutic agents and drugs targeting other cells of the tumor micro-environment. Angiogenesis inhibitors may be used as either monotherapy or in combination with other anticancer drugs. In this context, many preclinical and clinical studies revealed higher therapeutic effectiveness of the combined treatments compared with individual treatments. The proper understanding of synergistic treatment modalities of angiogenesis inhibitors as well as their wide range of cellular targets could provide effective tools for future therapies of many types of cancer.

Two major processes of blood vessel formation are implicated in the development of vascular system: vasculogenesis and angiogenesis. Vasculogenesis prevails in the embryo and refers to the formation ofde novo blood vessels by in situ differentiation of the mesoderm-derived angioblasts and endothelial precursors. Angiogenesis is the formation of new capillaries from pre-existing vessels and circulating endothelial precursors (Polverini, 2002; Chung et al., 2010; Ribatti and Djonov, 2012). Angiogenesis is a tightly controlled dynamic process that can occur physiologically in those tissues that undergo active remodeling in response to stress and hypoxia (Carmeliet, 2003; Folkman, 2007). However, it can be aberrantly activated during many pathological conditions such as cancer, diabetic retinopathy as well as numerous ischemic, inflammatory, infectious and immune disorders (Carmeliet, 2003; Ali and El-Remessy, 2009; Willis et al., 2011). Although the concept of proposing angiogenesis inhibitors as anticancer drugs received considerable skepticism when first presented by Dr. Folkman in the early 1970s (Folkman, 1971), active research in the field and subsequent clinical trials eventually resulted in US Food and Drug Administration (FDA) approval of bevacizumab for colorectal cancer in 2004 (Cohen et al., 2007). Since then, several angiogenic inhibitors have been identified. This review will provide an overview of the key mechanisms involved in tumor angiogenesis, classification of angiogenesis inhibitors as well as treatment rationales from the mechanistic point of view.

Sustained angiogenesis as a hallmark of cancer

Proliferating tumours tend to activate an angiogenic phenotype to fulfil their increased demand of oxygen and nutrients (Hanahan and Folkman, 1996; Carmeliet, 2005). Additionally, paracrine release of anti-apoptotic factors from activated endothelial cells in the newly formed vasculature supplies tumour cells with a survival privilege (Folkman, 2003). Consequently, in order to progress, tumors tend to activate an event called ‘angiogenic switch’ by shifting the balance of endogenous angiogenesis inducers and inhibitors towards a pro-angiogenic outcome. As a result, dormant lesion progresses into outgrowing vascularized tumor and eventually into a malignant phenotype (Hanahan and Folkman, 1996; Baeriswyl and Christofori, 2009). Hypoxia drives such imbalance through up-regulation of the transcription factor hypoxia inducible factor-1α (HIF-1α), which in turn increases the expression of many angiogenesis inducers as well as suppresses the expression of endogenous angiogenesis inhibitors (Pugh and Ratcliffe, 2003). In spite of that, accumulating evidence indicates that angiogenic cascade can be also driven by alternative HIF-1-independent pathways (Mizukami et al., 2007; Arany et al., 2008; Lee, 2013).

As summarized in Table 1, the angiogenesis inducers are a wide range of mediators that include many growth factors, a plethora of cytokines, bioactive lipids, matrix-degrading enzymes and a number of small molecules (Folkman, 1995; Folkman, 2003; Lopez-Lopez et al., 2004; Bouis et al., 2006; El-Remessy et al., 2007; Bid et al., 2011; MacLauchlan et al., 2011; Murakami, 2011; Fagiani and Christofori, 2013; Qin et al., 2013). Pro-angiogenic growth factors mostly activate a series of surface receptors in a series of paracrine and autocrine loops with the VEGF-A signaling representing the critical rate-limiting step, physiologically and pathologically. VEGF-A (traditionally known as VEGF) is the most potent VEGF isoform that acts mainly on VEGF receptor 2 (VEGFR2) to mediate vascular permeability, endothelial proliferation, migration and survival (Takahashi and Shibuya, 2005; Bouis et al., 2006). In spite of the well-established master roles of VEGF signaling in literature, those processes are probably accomplished through a highly regulated interplay between VEGF and other pro-angiogenic factors. In this context, basic fibroblast growth factor (bFGF) activation of the endothelium is required for maintenance of VEGFR2 expression and the ability to respond to VEGF stimulation (Murakami et al., 2011). Similarly, sphingosine-1-phosphate (S1P), a pleiotropic bioactive lipid that can directly contribute to tumor angiogenesis (reviewed in Sabbadini, 2011), is needed for VEGF-induced blood vessel formation, indicating the cooperation between S1P and VEGF in tumor angiogenesis (Visentin et al., 2006). As a net result, the pro-angiogenic interplay of those ligands and others dominates over the activities of two dozen endogenous angiogenesis inhibitors that can be either matrix-derived inhibitors or non–matrix-derived inhibitors (Nyberg et al., 2005).

Table 1. Pro-angiogenic mediators implicated in tumor angiogenesis

Category Examples References
Growth factors VEGFs Bouis et al., 2006
FGFs Ibid
TGFs Ibid
PDGFs Ibid
Insulin-like growth factors Lopez-Lopez et al., 2004; Bid et al., 2011
ANGs Fagiani and Christofori, 2013
Cytokines IL-8 Strieter et al., 2004
CSF-1 Lin et al., 2006
Bioactive lipids PGE2 Wang and Dubois, 2010
S1P Murakami, 2011
Matrix-degrading enzymes MMPs Bourboulia and Stetler-Stevenson, 2010
Heparanases Vlodavsky and Friedmann, 2001
Small mediators NO MacLauchlan et al., 2011
Peroxynitrite El-Remessy et al., 2007
Serotonin Qin et al., 2013
Histamine Qin et al., 2013

The multistep angiogenic process starts with vasodilation and increased permeability of existing vessels in response to tumor cell-secreted VEGF. This is accompanied by loosening of pericytes covering mediated by angiopoietin-2 (ANG2), a ligand of tyrosine kinase with immunoglobulin-like and EGF-like domains 2 (TIE2) receptor (Bergers and Benjamin, 2003; Jain, 2003; Fagiani and Christofori, 2013). Meanwhile, many secreted matrix-degrading enzymes, such as MMPs and heparanases, function in concert to dissolve the basement membrane and to remodel the extracellular matrix (ECM) as well as to liberate more pro-angiogenic growth factors (bFGF and VEGF) from matrix heparan sulfate proteoglycans (HSPGs) respectively (Houck et al., 1992; Whitelock et al., 1996; Vlodavsky and Friedmann, 2001; Tang et al., 2005; van Hinsbergh and Koolwijk, 2008). The overall chemotactic angiogenic stimuli guide endothelial cells to migrate, to align into tube-like structures and to eventually form new blood vessels. However, such blood vessels are characterized by being disorganized, chaotic, hemorrhagic and poorly functioning (Bergers and Benjamin, 2003).

The angiogenic phenotype in tumor micro-environment can further be sustained and extravagated by the recruitment of other types of stromal cells. Stromal cells such as fibroblasts, mesenchymal stem cells and various bone marrow-derived myeloid cells including macrophages, TIE2-expressing monocytes, neutrophils and mast cells contribute to tumor angiogenesis through their production of growth factors, cytokines and proteases (Murdoch et al., 2008; Joyce and Pollard, 2009; Cirri and Chiarugi, 2011). For example, in response to cancer cell-derived TGF-β, PDGF or bFGF, fibroblasts are transformed to an activated phenotype with a higher proliferative activity and myofibroblastic characteristics (Kalluri and Zeisberg, 2006; Cirri and Chiarugi, 2011). Such carcinoma-associated fibroblasts (CAFs) were shown to promote angiogenesis and metastasis by secreting large amounts of MMP-2 and MMP-9 as well as by expressing many cytokines and chemokines that resulted in immune cell infiltration (Gerber et al., 2009; Giannoni et al., 2010). Furthermore, it has been shown that PDGF-C produced by CAFs is able to elicit VEGF production from tumor cells, thereby sustaining the angiogenic shift (Crawford et al., 2009). Similarly, tumor-associated macrophages (TAMs), one of the bone marrow myeloid-derived cells, are induced to develop into polarized type II (alternatively activated or M2 macrophages), upon exposure to tumor hypoxia and tumor cell-derived cytokines (Leek et al., 2002; Rogers and Holen, 2011). M2 macrophages tend to produce many pro-angiogenic growth factors, cytokines and matrix-degrading enzymes such as VEGF, PDGF, bFGF, TNF-α, COX-2, MMP-9, MMP-7 and MMP-12 (Lewis and Pollard, 2006).

From another perspective, angiogenesis may be dispensable for progression of some malignancies. For example, some tumours may co-opt pre-existent vessels as an alternative way to obtain blood supply. Vessel co-option was first described in the brain, one of the most densely vascularized organs, in which tumours may develop in earlier stages without the activation of angiogenic response (Holashet al., 1999; Leenders et al., 2002; Bergers and Benjamin, 2003; Hillen and Griffioen, 2007). In another example, hypovascularized tumors such as pancreatic ductal adenocarcinoma may involve certain adaptation to flourish in the absence of prominent angiogenesis (Bergers and Hanahan, 2008). Obviously, in both cases, tumors may be intrinsically indifferent to angiogenesis inhibitors. However, in most other cases, therapy directed towards the vasculature of solid tumors is being considered as an important direction in cancer treatment.

Classification of angiogenesis inhibitors

Growth of newly formed vessels in tumor micro-environment can be inhibited directly by targeting endothelial cells in the growing vasculature or indirectly by targeting either tumor cells or the other tumor-associated stromal cells. Therefore, angiogenesis inhibitors can be classified into direct and indirect inhibitors (Kerbel and Folkman, 2002; Folkman, 2007).

Direct endogenous inhibitors of angiogenesis

Direct endogenous inhibitors of angiogenesis, such as angiostatin, endostatin, arrestin, canstatin, tumstatin and others, are fragments released on proteolysis of distinct ECM molecules. Endogenous inhibitors prevent vascular endothelial cells from proliferating, migrating in response to a spectrum of angiogenesis inducers, including VEGF, bFGF, IL-8 and PDGF (Kerbel and Folkman, 2002; Abdollahi et al., 2004; Mundel and Kalluri, 2007; Ribatti, 2009). This direct anti-angiogenic effect may be mediated by interference with endothelial integrins along with several intracellular signaling pathways (Mundel and Kalluri, 2007). For example, the ability of tumstatin-derived active peptide to inhibit angiogenesis and tumour growth is associated with the expression of the adhesion receptor, αvβ3 integrin, on tumor endothelial cells (Eikesdal et al., 2008). Through binding αvβ3 integrin, full tumstatin was found to inhibit endothelial cell activation of focal adhesion kinase, PI3K, Akt, mammalian target of rapamycin (mTOR) and others (Maeshima et al., 2002). Direct targeting of those signaling pathways by endogenous inhibitors was thought to be the least likely to induce acquired drug resistance because they target endothelial cells with assumed genetic stability rather than unstable mutating tumour cells (Kerbel and Folkman, 2002). However, endostatin has not yet led to any documented benefit to patients in randomized phase III trials, or even modest activity in phase II trials (Ellis and Hicklin, 2008).

Indirect inhibitors of angiogenesis

Indirect inhibitors of angiogenesis classically prevent the expression or block the activity of pro-angiogenic proteins (Folkman, 2007). For example, Iressa, an EGF receptor (EGFR) TK inhibitor (TKI), blocks tumour expression of many pro-angiogenic factors; bevacizumab, a monoclonal antibody, neutralizes VEGF after its secretion from tumour cells whereas sunitinib, a multiple receptor TKI, blocks the endothelial cell receptors (VEGFR1, VEGFR2 and VEGFR3), preventing their response to the secreted VEGF (Folkman, 2007; Roskoski, 2007). In addition, this class extends to include conventional chemotherapeutic agents, targeted therapy against oncogenes and drugs targeting other cells of the tumor micro-environment (Kerbel et al., 2000; Ferrara and Kerbel, 2005).

Conventional chemotherapeutic agents

Conventional chemotherapeutic agents have been shown to have anti-angiogenic properties in addition to the ability to induce direct cancer cell death. Such chemotherapeutic agents can affect the endothelial cell population in the tumour bed during treatment cycles because they have significantly higher proliferation rates than resting endothelium outside a tumor, making them more susceptible to cytotoxic effect (Kerbel et al., 2000; Folkman, 2003). However, the cyclic treatment rationale of cytotoxic drugs allows the potential damage to the tumour vasculature to be repaired during the long breaks. Thus, continuous low doses of chemotherapeutic agents were suggested as a way to reduce side effects and drug resistance (Drevs et al., 2004). This modality is termed metronomic therapy, and clinically, it refers to the daily administration of 5–10% of the phase II-recommended dose of the chemotherapeutic agent (Penel et al., 2012). The extended use of such low doses of cytotoxic agents elicits an anti-angiogenic activity through induction of endothelial cell apoptosis and decreasing the level of circulating endothelial precursors (Hamano et al., 2004; Shahrzad et al., 2008). In clinical investigations, metronomic dosing of cyclophosphamide and others showed promising efficacy in patients with advanced, multiple metastasized and/or multiple pretreated solid tumours (Lord et al., 2007; Fontana et al., 2010; Nelius et al., 2011; Gebbia et al., 2012; Briasoulis et al., 2013; Navid et al., 2013).

VEGF-targeted therapy

VEGF-targeted therapy includes neutralizing antibodies to VEGF (e.g. bevacizumab) or VEGFRs (e.g. ramucirumab), soluble VEGFR/VEGFR hybrids (e.g. VEGF-Trap) and TKIs with selectivity for VEGFRs (e.g. sunitinib and sorafenib; Baka et al., 2006; Ellis and Hicklin, 2008; Hsu and Wakelee, 2009). Bevacizumab, a humanized monoclonal antibody against all isoforms of VEGF-A, has been approved for the treatment of colorectal, lung, glioblastoma and renal cell carcinoma (Hsu and Wakelee, 2009). Many other clinical trials with promising efficacy were also conducted in other cancers such as head and neck cancer, hepatocellular carcinoma, ovarian cancer, metastatic melanoma and gastric cancer (Argiris et al., 2011; 2013; Burger et al., 2011; Ohtsu et al., 2011; Fang et al., 2012; Minor, 2012; Schuster et al., 2012; Van Cutsem et al., 2012). However, for metastatic breast cancer, bevacizumab had been initially granted an accelerated FDA approval, which was later withdrawn due to lack of improvement evidence in disease-related symptoms or overall survival (Burstein, 2011; Montero et al., 2012). Similarly, clinical trials showed that the addition of bevacizumab to the treatment regimens of advanced pancreatic cancer did not extend overall survival (Chiu and Yau, 2012). The neutralization of VEGF-A can also be achieved by soluble receptor construct (VEGF-Trap) that monomerically ‘traps’ the different isoforms of VEGF-A, in addition to VEGF-B and placental growth factor (Rudge et al., 2007). VEGF-Trap showed clinical benefit in a phase III trial of oxaliplatin pretreated metastatic patients with colorectal cancer, and is currently being investigated in a prostate cancer phase III trial (Gaya and Tse, 2012). TKIs are small molecules with different chemical structures that have the ability to interact physically with the highly conserved kinase domain shared by different VEGFRs as well as PDGF receptors (PDGFRs), FGF receptors (FGFRs), EGFR, Raf kinases and c-Kit (a receptor of the pluripotent cell growth factor, stem cell factor). Such interaction directly inhibits tyrosine phosphorylation and the subsequent many downstream pro-angiogenic signaling networks (Baka et al., 2006; Ivy et al., 2009). Those multi-targeted TKIs demonstrated efficacy against various solid malignancies in different clinical trials, some of which have lead eventually to FDA approval of sunitinib and sorafenib. Sunitinib, known to inhibit several receptor TKs (RTKs) including VEGFR1–3, PDGFR-α, PDGFR-β, c-Kit, colony-stimulating factor-1 receptor (CSF-1R) and Flt-3, was approved for the treatment of renal cell carcinoma and gastrointestinal stromal cell tumours. Sorafenib that acts also by inhibiting VEGFR1–3 and PDGFR-β in addition to the serine–threonine kinases Raf-1, B-Raf, was approved for hepatocellular carcinoma in addition to renal cell carcinoma (Llovet et al., 2008; Ivy et al., 2009; Huang et al., 2010).

FGF-targeted therapies

FGF-targeted therapies were recently reconsidered as promising anti-angiogenic and anti-tumor agents after a long period of little attention for drug development, partly due to redundancy (Bono et al., 2013). The FGFR superfamily with its 18 ligands and four receptors has been involved in endothelial cell migration, proliferation and differentiation (Presta et al., 2005). Therapeutic targeting of FGF/FGFR signalling was accomplished by either monoclonal antibodies that inhibit FGFs binding, small molecules that inhibit FGFR TK activity or allosteric modulators that bind the extracellular FGFR domain. Monoclonal antibodies against bFGF displayed potent anti-tumor and anti-angiogenic effects in different preclinical cancer models, which warrant further clinical evaluation (Zhao et al., 2010; Wang et al., 2012). Pan inhibitors of the FGFR TKs such as AZD4547 (blocks the activity of FGFR1–3) and ponatinib (blocks all the FGFR isoforms) elicited potent anti-tumor activities in preclinical investigations so they are currently being evaluated in clinical trials. Those inhibitors displayed the greatest potency in FGFR-driven cancer models, which may be attributed to the interference with the oncogenic functions of either amplified or constitutively active FGFR (Dutt et al., 2011; Zhao et al., 2011; Gavine et al., 2012; Gozgit et al., 2012). Accordingly, further studies are needed to evaluate the relative contribution of angiogenic versus oncogenic inhibitory mechanisms towards the overall anti-tumor activity. The allosteric antagonist of the FGFR, SSR128129E, showed a strong anti-angiogenic activity in addition to tumour growth and metastasis inhibitory effects in animal models of arthritis and cancer respectively. Because allosteric modulators leave a residual level of baseline signalling, they have the ability to fine-tune target biological responses. As a result, allosteric multi-FGFR inhibitors may have an improved benefit/risk ratio that is not attainable with the other TKIs (Bonoet al., 2013; Herbert et al., 2013). However, preclinical findings suggest that long-term clinical outcomes may improve with blockade of additional pro-angiogenic RTKs that may also reduce the risk of drug resistance (Hilberg et al., 2008). For example, dual inhibition of VEGFRs and FGFRs using brivanib produced enduring tumour stasis and angiogenic blockade following the failure of VEGF-targeted therapies (Allen et al., 2011). Furthermore, triple inhibition of FGFRs, VEGFRs and PDGFR(s) using dovitinib (TKI258) or nintedanib (BIBF 1120) displayed broad-spectrum anti-tumour activities in several tumour xenograft models as well as promising data in clinical trials. Combined inhibition of FGFR/VEGFR/PDGFR targets not only tumour cells, but also endothelial cells, pericytes and smooth muscle cells, resulting in an effective inhibition of tumour growth, angiogenesis and metastasis even in advanced tumour stages (Hilberg et al., 2008; Ledermann et al., 2011; Taeger et al., 2011; Chenet al., 2012; Angevin et al., 2013).

Oncogene-targeted therapy

Oncogenes, genes that cause the transformation of normal cells into cancerous cells, are thought to up-regulate many pro-angiogenic proteins. Therefore, anticancer drugs that were developed for their capacity to block an oncogene also have an indirect anti-angiogenic activity (Kerbel et al., 2000; Bergers and Benjamin, 2003; Folkman, 2003). For example, dasatinib and other inhibitors of sarcoma (Src), an aberrantly activated non-RTK associated with many human malignancies, showed potent anti-angiogenic effects through the down-regulation of VEGF and IL-8 (Summy et al., 2005; Han et al., 2006; Haura et al., 2010). Another example is to target the oncogenic Ras using farnesyl transferase (FT) inhibitors, which inhibit post-translational farnesylation of Ras that governs the latter’s activity (Awada et al., 2002). FT inhibitors were found to inhibit tumor VEGF expression and block FTase-dependent Ras activation, which is critically involved in VEGF-elicited angiogenic signal transduction and angiogenesis (Han et al., 2005; Izbicka et al., 2005; Kim et al., 2010). In addition to classical oncogenes inhibition, interference with other tumor-deregulated signaling pathways would offer another approach in targeting angiogenesis. For example, inhibitors of heat shock protein 90 (HSP90), a chaperone molecule known to protect oncoproteins from misfolding and degradation in the protein-rich intracellular environment, were found to prevent VEGF production and to disrupt multiple pro-angiogenic signalling pathways in numerous cancer cells. They were also shown to inhibit tumour growth and vascularity of different human tumor xenografts (Sanderson et al., 2006; Langet al., 2007; Eccles et al., 2008; Trepel et al., 2010; Moser et al., 2012). Proteasome inhibitors, such as bortezomib (PS-341) or MG-132, were also shown to reduce tumour growth and vascularity of squamous cell carcinoma and pancreatic cancer xenograft probably through inhibition of NF–κB-dependent release of pro-angiogenic gene products, VEGF and IL-8 (Sunwoo et al., 2001; Nawrocki et al., 2002; Matsuo et al., 2009). Similarly, inhibition of B-cell lymphoma 2 (Bcl-2), a prosurvival protein that regulates apoptosis by preventing the mitochondrial release of pro-apoptogenic factors, was shown to prevent NF-κB-mediated release of the pro-angiogenic factors IL-8 and CXC chemokine ligand 1 (CXCL1) as well as VEGF in tumor-associated endothelial cells and pancreatic cell lines respectively (Karl et al., 2005; Wang et al., 2008). Moreover, (−)-gossypol, a natural BH3 mimetic that inhibits BH3 domain of Bcl-2 as well as related prosurvival proteins (Bcl-xL and Mcl-1), was shown to remarkably decrease microvessel density in human prostate tumour PC-3 xenografts through decrease of VEGF and IL-8 release as well as blocking multiple steps in VEGF-activated biological events (Karaca et al., 2008; Pang et al., 2011).

Matrix degrading and remodelling-targeted therapy

Matrix degrading and remodelling are activated by tumors to modify local micro-environment, which in turn promote their angiogenic potential (Bergers et al., 2000; Vlodavsky and Friedmann, 2001). Up-regulation of expression and activity of several endogenous MMPs including MMP-2, MMP-9 as well as MMP-3 and MMP-7 have been identified in invasive tumors (for a review, see Bourboulia and Stetler-Stevenson, 2010). Consequently, inhibitors of MMPs were extensively pursued as a therapeutic strategy for treating cancer. Unfortunately, MMPs intervention strategies had met with limited clinical success because of severe toxicities and associated metastasis-promoting effect (Coussens et al., 2002; Devy et al., 2009). Furthermore, the paradoxical roles of tissue inhibitors of metalloproteinases (TIMPs) may contribute to such failure depending on the net balance of TIMPs and MMPs in tumour stroma (Jiang et al., 2002). As a result, efforts were directed at therapies exploiting endogenous MMP inhibitors, TIMPs or monoclonal antibodies against individual MMPs (Martens et al., 2007; Jarvelainen et al., 2009). For example, DX-2400, a highly selective fully human MMP-14 inhibitory antibody, was found to block pro-MMP-2 processing on tumor and endothelial cells, inhibited angiogenesis, and slowed tumor progression and formation of metastatic lesions (Devy et al., 2009). Alternatively, in order to reduce toxicity and enhance drug delivery, polymeric nanoparticulate delivery systems could be used to target individual components of ECM. For example, targeted delivery of antisense inhibitors of laminin-8, a vascular basement membrane component, by conjugation to the natural drug carrier β-poly(L-malic acid) significantly reduced tumour microvessel density and increased animal survival in an experimental model of glioblastoma (Fujita et al., 2006). Similarly, a nano delivery system that incorporate peptides against proteolytically processed type IV collagen significantly accumulated in tumors and blocked angiogenesis in experimental models (Mueller et al., 2009). However, the highly sulfated oligosaccharides, Heparan (HS) mimetics highly sulfated oligosaccharides, were shown to have a heparanase-inhibiting effect sequestering, in turn, many heparan sulfate proteoglycan (HSPG)-binding factors (Johnstone et al., 2010; Dredge et al., 2011). In preclinical studies, HS mimetics have effectively targeted multiple HSPG-dependent functions and have resulted in decreased in vivo tumor growth, tumor invasion, tumor metastasis and angiogenesis (Johnstone et al., 2010; Dredge et al., 2011; Zhou et al., 2011). Clinically, the heparanase inhibitor PI-88 showed preliminary efficacy as an adjunct therapy for post-operative hepatocellular carcinoma (Liu et al., 2009).

Tumour-associated stromal cell-targeted therapy

Tumour-associated stromal cells crosstalk is a perquisite for the formation of a tumour vasculature, an essential step for tumour progression (Lorusso and Ruegg, 2008). Interference with those crosstalk circuits through intervention of cellular adhesion (highlighted in next paragraph) or tumor-induced recruitment of different stromal cells may be considered as an indirect way of anti-angiogenic therapy (Ferrara and Kerbel, 2005). The latter can be supported by studies in which inhibition of macrophage infiltration, for example, by either genetic ablation of the macrophage CSF-1 or liposomal clodronate-induced macrophage depletion, was shown to delay the angiogenic switch and malignant transition (Giraudo et al., 2004; Lin et al., 2006). Furthermore, CSF-1R kinase inhibitors were found to reduce tumor-associated vascularity in two different tumor mouse models (Kubota et al., 2009; Mantheyet al., 2009). In addition, clodronate and other related bisphosphonates, originally used to treat skeletal complications in patients with tumour-induced osteolysis, were shown to exert potent anti-tumour and anti-angiogenic effects in many other studies (Fournier et al., 2002; Santini et al., 2003; Stathopoulos et al., 2008). Zoledronic acid, a third-generation bisphosphonate, was also found to reduce a number of tumour-associated macrophages and shift their phenotype from M2 to M1, resulting in a reduction in TAM-associated production of VEGF in murine models of spontaneous mammary carcinogenesis and mesothelioma (Coscia et al., 2010; Veltman et al., 2010). Clinically, repeated low-dose therapy with zoledronic acid, which maintains active drug plasma concentration, was able to induce an early remarkable and long-lasting decrease of VEGF levels in patients with cancer (Santini et al., 2007). In another example, inhibition of mobilization of neutrophils, from bone marrow and their infiltration into tumour, using neutralizing anti–prokineticin-2, an antibody against a secreted protein known also as BV8, was shown to impair the initial angiogenic switch in a multistage pancreatic beta cell tumorigenesis model (Shojaei et al., 2008). Furthermore, the neutralizing anti-BV8 was found to prevent myeloid cell-dependent tumour angiogenesis in several xenograft models (Shojaei et al., 2007). Cancer-associated fibroblasts (CAF) can also be targeted with thapsigargin analogue coupled with peptides specific for fibroblast activation protein (FAP), a CAF membrane-bound protease whose catalytic site has access to the peritumoural fluid of the tumor micro-environment. This extracellular activation results in the death of CAFs as well as pericytes and endothelial cells within milieu of different human tumor xenografts (Brennen et al., 2012).

Cell adhesion molecules (CAMs)-targeted therapy

CAMs are cell surface proteins known to be involved in binding with other counter-receptors on adjacent cells or surrounding ECM macromolecules (Aplin et al., 1998). Many CAMs, such as αv-integrins, E-selectin, N-cadherin and VE-cadherin, have been implicated in tumour angiogenesis (Bischoff, 1997; Tei et al., 2002; Nakashima et al., 2003; Weis and Cheresh, 2011). For example, αv-integrins are expressed on surface of endothelial cells and can determine whether cells can adhere to and survive in a particular micro-environment. A number of matrix-derived fragments have the ability to act as endogenous angiogenesis inhibitors through binding to integrins on endothelial cells, disrupting physical connections and suppressing signalling events associated with cell survival, migration and proliferation (Nyberg et al., 2005). Consequently, integrins antagonism using peptidomimetics (e.g. cilengitide), monoclonal antibodies (e.g. volociximab) or oral small-molecule compounds have been investigated in a wide range of malignancies (Huveneers et al., 2007). Cilengitide is a cyclized pentapeptide peptidomimetic designed to compete for the arginine-glycine-aspartic acid (RGD) peptide sequence, thereby blocking the ligation of the αvβ3 and αvβ5 integrins to matrix proteins (Hariharan et al., 2007). Cilengitide is mainly under clinical development for glioblastoma; however, clinical trials of other malignancies such as head and neck cancer as well as lung cancer were also initiated (Reardon and Cheresh, 2011; Vermorken et al., 2012; Manegold et al., 2013). Alternatively, cyclic peptides containing RGD motif could guide nanoparticulate delivery system, which incorporates anti-angiogenic cytotoxic agents such as doxorubicin, paclitaxel or combretastatin A4, to accumulate specifically in tumor vasculature with no overt systemic toxicity (Murphy et al., 2008; Ruoslahti et al., 2010; Wang et al., 2011). Volociximab, a chimeric humanized monoclonal antibody that selectively inhibits the αvβ1 integrin interaction with fibronectin, has been evaluated also in clinical trials for solid tumours such as renal cell carcinoma, recurrent ovarian cancer, advanced non–small-cell lung cancer and metastatic pancreatic cancer (Figlin et al., 2006; Evans et al., 2007; Jarvelainen et al., 2009; Vergote et al., 2009; Besse et al., 2013). Cadherins constitute a superfamily of molecules that mediate calcium-dependent cell–cell adhesions. The intracellular domains of cadherins directly bind to β-catenin and link with cytoskeletal components, providing the molecular basis for stable cell–cell adhesion (Zhang et al., 2010). Targeting cadherin signalling may also represent another way for tumor angiogenesis intervention. For example, ADH-1, a cyclic pentapeptide containing the cell adhesion recognition site (His-Ala-Val) required for N-cadherin adhesion, was shown to possess anti-angiogenic and anti-tumour activity (Blaschuk et al., 2005; Blaschuk, 2012). Similarly, monoclonal antibody directed against specific region of VE-cadherin was able to inhibit tumor angiogenesis and growth with no side effects on normal vasculature (Corada et al., 2002; May et al., 2005).

Inflammatory angiogenesis-targeted therapy

Targeting inflammatory angiogenesis, responsible for a substantial part of tumour vascularization initiated by infiltrating leukocytes, may be considered as another indirect anti-angiogenic strategy (Albini et al., 2005). Moreover, as mentioned before, tumour-infiltrating leukocytes contribute into malignant progression through production of many pro-inflammatory cytokines, chemokines and enzymes that can mostly induce angiogenic cascade (Balkwill et al., 2005). Such vital roles have been supported by the early observation that nonsteroidal anti-inflammatory drugs can inhibit tumour angiogenesis and, in turn, tumor progression (Albini et al., 2005). For example, ibuprofen was found to decrease tumor growth and metastatic potential in mice models through modulation of angiogenesis (Yao et al., 2005). Moreover, selective inhibitors of COX-2, an inducible enzyme that catalyses the production of prostanoids from arachidonic acid, were also shown to inhibit angiogenesis (Tsujii et al., 1998; Wei et al., 2004). The anti-angiogenic effect of COX-2 inhibitors may be contributed, in part, by decreasing the COX-2 metabolic product PGE2, the predominant PG in solid tumors known to stimulate cancer cells to produce pro-angiogenic factors such as VEGF and bFGF as well as many other factors belonging to CXC chemokines family (Strieter et al., 2004; Wang et al., 2006; Wang and Dubois, 2010). Members of the CXC chemokine family are heparin-binding proteins that possess disparate regulative roles in angiogenesis. For example, the ELR+ CXC chemokines, characterized by highly conserved three amino acid motifs (Glu-Leu-Arg; ‘ELR’ motif), are potent promoters of angiogenesis, whereas the IFN-inducible (ELR−) CXC chemokines are inhibitors of angiogenesis (Strieter et al., 2004). The use of repertaxin, originally designed to target the ELR+ CXC chemokine receptors CXCR1 and CXCR2 on neutrophils to prevent their migration to sites of inflammation, was found to inhibit tumor angiogenesis, thereby suppressing tumour progression in a genetic model of pancreatic ductal adenocarcinoma (Ijichi et al., 2011). It would be beneficial to explore other small-molecule CXCR2 antagonists that have already been developed for the treatment of inflammatory diseases in different preclinical models of cancer, especially inflammation-associated cancers (refer to Chapman et al., 2009 for a list of newly developed CXCR2 antagonists used in the treatment of inflammatory diseases of the lung).

Mechanisms of enhanced therapeutic efficacy

  • Dual targeting of tumor vasculature
  • Targeting different cell types of tumor micro-environment
  • Normalization of tumor vasculature
  • Chemosensitization of tumor cells
  • Interference with the repair of cytotoxic drug-induced damage and resistance mechanisms

Consequences of anti-angiogenic therapy with other anticancer therapy

  • Contrary to initial expectations, treatment with angiogenesis inhibitors was associated with unexpected toxicities. The toxicity profiles of those inhibitors reflect the systemic disturbance of growth factor signalling pathways that mediate their anti-angiogenic activity (Elice and Rodeghiero, 20102012). In this context, disturbance of the tight endothelial cell-platelet interaction that maintains vascular integrity results in bleeding complications, gastrointestinal perforations, and disturbed wound and ulcer healing (Verheul and Pinedo, 2007). In general, the incidence of those adverse effects increases when anti-angiogenic agent is combined with chemotherapy. For example, bleeding complications have been observed in patients with colorectal cancer treated with chemotherapy in combination with bevacizumab (Kabbinavar et al., 2003; Giantonio et al., 2006). In non–small-cell lung cancer, some patients treated with bevacizumab in combination with carboplatin and paclitaxel experienced severe or fatal pulmonary haemorrhage (Johnson et al., 2004). Furthermore, a higher incidence of gastrointestinal perforation was observed in patients with colorectal cancer given bevacizumab in combination with chemotherapy compared with chemotherapy alone (Hurwitz et al., 2004). Similarly, thrombotic events have been observed in patients treated with angiogenesis inhibitors, especially when these agents are given in combination with chemotherapy (Verheul and Pinedo, 2007). Treatment of patients with cancer with angiogenesis inhibitors is frequently associated with hypertension, which may require the addition of regular anti-hypertensive agent (Izzedine et al., 2009).

Summary and future directions

  • Angiogenesis is a critical process that occurs pathologically in many malignancies due to changing balance of endogenous angiogenesis inducers and inhibitors, leading to the activation of nearby endothelial cells to form new vasculature. Consequently, angiogenesis can be targeted to restrict initiation, growth and progression of most of angiogenesis-dependent malignancies. Numerous angiogenic inhibitors have been identified, some of which are currently being investigated in clinical trials and some others were even approved for cancer therapies. These angiogenesis inhibitors were classified based on their target into two main classes: direct and indirect inhibitors. Indirect angiogenesis inhibitors can be further subclassified based on their interference mechanisms with the angiogenic cascade. A list of major categories and molecular targets for angiogenesis inhibitors is shown in Table 2.
  • Most angiogenesis inhibitors conferred clinical benefits mainly when combined with other chemotherapeutic/targeted therapies rather than being used as monotherapy. Unfortunately, many anti-angiogenic agents were shown to be associated with overt systemic toxicity as well as resistance emergence and disease recurrence. Drug resistance in anti-angiogenic therapy may result from a plethora of pro-angiogenic factors released by inappropriately functioning host cells in the tumor micro-environment as a compensatory mechanism. Therefore, the strategy of targeting endothelial cells alone may not be enough as explained in the previous texts, requiring the proposal of different rationales in which other cellular compartments of tumor micro-environment are targeted to attain proper anti-angiogenic and anti-tumor response. That highlights the importance of considering tumor micro-environment as a dynamic system, as depicted in Figure 1 in which interference with any of its components may be an approach to interfere with cancer hallmarks, including angiogenesis.

9.5.5 LUCITANIB a VEGFR/FGFR dual kinase inhibitor in Phase 2 trials

Dr.  Anthony Melvin Crasto

source: http://medcheminternational.blogspot.com/2015/01/lucitanib-vegfrfgfr-dual-kinase.html

Lucitanib.png
LUCITANIB
6-[7-[(1-aminocyclopropyl)methoxy]-6-methoxyquinolin-4-yl]oxy-N-methylnaphthalene-1-carboxamide
6-(7-((l-aminocyclopropyl)methoxy)-6-methoxyquinolin-4-yloxy)- N-methyl- 1 -naphthamide
1058137-23-7 (E-3810 free base); 1058137-84-0  (E-3810 HCl salt)
E-3810, E-3810 amine, UNII-PP449XA4BH, E3810, Lucitanib [INN], AL3810
Molecular Formula:C26H25N3O4
Molecular Weight:443.4944 g/mol
PATENT SUBMITTED GRANTED
Spiro Substituted Compounds As Angiogenesis Inhibitors [US8163923] 2008-09-18 2012-04-24
A 4-(3-methoxypropoxy)-3-methylpyridinyl derivative of timoprazole that is used in the therapy of STOMACH ULCERS and ZOLLINGER-ELLISON SYNDROME. The drug inhibits H(+)-K(+)-EXCHANGING ATPASE which is found in GASTRIC PARIETAL CELLS.
For in advanced solid tumors.
Lucitanib (E-3810): Lucitanib, also known as E-3810,  is a novel dual inhibitor targeting human vascular endothelial growth factor receptors (VEGFRs) and fibroblast growth factor receptors (FGFRs) with antiangiogenic activity. VEGFR/FGFR dual kinase inhibitor E-3810 inhibits VEGFR-1, -2, -3 and FGFR-1, -2 kinases in the nM range, which may result in the inhibition of tumor angiogenesis and tumor cell proliferation, and the induction of tumor cell death. Both VEGFRs and FGFRs belong to the family of receptor tyrosine kinases that may be upregulated in various tumor cell type
Lucitanib (E-3810) Structure

Overview

Lucitanib is an oral, potent inhibitor of the tyrosine kinase activity of fibroblast growth factor receptors 1 through 3 (FGFR1-3), vascular endothelial growth factor receptors 1 through 3 (VEGFR1-3) and platelet-derived growth factor receptors alpha and beta (PDGFR α-ß). We own exclusive development and commercial rights to lucitanib on a global basis, excluding China. Lucitanib rights to markets outside of the U.S. and Japan have been sublicensed to Les Laboratoires Servier (Servier). We are collaborating with Servier on the global clinical development of lucitanib.

Read Full Post »

Hypoxia Inducible Factor 1 (HIF-1)

Writer and Curator: Larry H Bernstein, MD, FCAP

7.9  Hypoxia Inducible Factor 1 (HIF-1)

7.9.1 Hypoxia and mitochondrial oxidative metabolism

7.9.2 Hypoxia promotes isocitrate dehydrogenase-dependent carboxylation of α-ketoglutarate to citrate to support cell growth and viability

7.9.3 Hypoxia-Inducible Factors in Physiology and Medicine

7.9.4 Hypoxia-inducible factor 1. Regulator of mitochondrial metabolism and mediator of ischemic preconditioning

7.9.5 Regulation of cancer cell metabolism by hypoxia-inducible factor 1

7.9.6 Coming up for air. HIF-1 and mitochondrial oxygen consumption

7.9.7 HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption

7.9.8 HIF-1. upstream and downstream of cancer metabolism

7.9.9 In Vivo HIF-Mediated Reductive Carboxylation

7.9.10 Evaluation of HIF-1 inhibitors as anticancer agents

 

 

7.9.1 Hypoxia and mitochondrial oxidative metabolism

Solaini G1Baracca ALenaz GSgarbi G.
Biochim Biophys Acta. 2010 Jun-Jul; 1797(6-7):1171-7
http://dx.doi.org/10.1016/j.bbabio.2010.02.011

It is now clear that mitochondrial defects are associated with a large variety of clinical phenotypes. This is the result of the mitochondria’s central role in energy production, reactive oxygen species homeostasis, and cell death. These processes are interdependent and may occur under various stressing conditions, among which low oxygen levels (hypoxia) are certainly prominent. Cells exposed to hypoxia respond acutely with endogenous metabolites and proteins promptly regulating metabolic pathways, but if low oxygen levels are prolonged, cells activate adapting mechanisms, the master switch being the hypoxia-inducible factor 1 (HIF-1). Activation of this factor is strictly bound to the mitochondrial function, which in turn is related with the oxygen level. Therefore in hypoxia, mitochondria act as [O2] sensors, convey signals to HIF-1directly or indirectly, and contribute to the cell redox potential, ion homeostasis, and energy production. Although over the last two decades cellular responses to low oxygen tension have been studied extensively, mechanisms underlying these functions are still indefinite. Here we review current knowledge of the mitochondrial role in hypoxia, focusing mainly on their role in cellular energy and reactive oxygen species homeostasis in relation with HIF-1 stabilization. In addition, we address the involvement of HIF-1 and the inhibitor protein of F1F0 ATPase in the hypoxia-induced mitochondrial autophagy.

Over the last two decades a defective mitochondrial function associated with hypoxia has been invoked in many diverse complex disorders, such as type 2 diabetes [1] and [2], Alzheimer’s disease [3] and [4], cardiac ischemia/reperfusion injury [5] and [6], tissue inflammation [7], and cancer [8][9][10],[11] and [12].

The [O2] in air-saturated aqueous buffer at 37 °C is approx. 200 μM [13]; however, mitochondria in vivo are exposed to a considerably lower [O2] that varies with tissue and physiological state. Under physiological conditions, most human resting cells experience some 5% oxygen tension, however the [O2] gradient occurring between the extracellular environment and mitochondria, where oxygen is consumed by cytochrome c oxidase, results in a significantly lower [O2] exposition of mitochondria. Below this oxygen level, most mammalian tissues are exposed to hypoxic conditions  [14]. These may arise in normal development, or as a consequence of pathophysiological conditions where there is a reduced oxygen supply due to a respiratory insufficiency or to a defective vasculature. Such conditions include inflammatory diseases, diabetes, ischemic disorders (cerebral or cardiovascular), and solid tumors. Mitochondria consume the greatest amount (some 85–90%) of oxygen in cells to allow oxidative phosphorylation (OXPHOS), which is the primary metabolic pathway for ATP production. Therefore hypoxia will hamper this metabolic pathway, and if the oxygen level is very low, insufficient ATP availability might result in cell death [15].

When cells are exposed to an atmosphere with reduced oxygen concentration, cells readily “respond” by inducing adaptive reactions for their survival through the AMP-activated protein kinase (AMPK) pathway (see for a recent review [16]) which inter alia increases glycolysis driven by enhanced catalytic efficiency of some enzymes, including phosphofructokinase-1 and pyruvate kinase (of note, this oxidative flux is thermodynamically allowed due to both reduced phosphorylation potential [ATP]/([ADP][Pi]) and the physiological redox state of the cell). However, this is particularly efficient only in the short term, therefore cells respond to prolonged hypoxia also by stimulation of hypoxia-inducible factors (HIFs: HIF-1 being the mostly studied), which are heterodimeric transcription factors composed of α and β subunits, first described by Semenza and Wang [17]. These HIFs in the presence of hypoxic oxygen levels are activated through a complex mechanism in which the oxygen tension is critical (see below). Afterwards HIFs bind to hypoxia-responsive elements, activating the transcription of more than two hundred genes that allow cells to adapt to the hypoxic environment [18] and [19].

Several excellent reviews appeared in the last few years describing the array of changes induced by oxygen deficiency in both isolated cells and animal tissues. In in vivo models, a coordinated regulation of tissue perfusion through vasoactive molecules such as nitric oxide and the action of carotid bodies rapidly respond to changes in oxygen demand [20][21][22][23] and [24]. Within isolated cells, hypoxia induces significant metabolic changes due to both variation of metabolites level and activation/inhibition of enzymes and transporters; the most important intracellular effects induced by different pathways are expertly described elsewhere (for recent reviews, see [25][26] and [27]). It is reasonable to suppose that the type of cells and both the severity and duration of hypoxia may determine which pathways are activated/depressed and their timing of onset [3][6][10][12][23] and [28]. These pathways will eventually lead to preferential translation of key proteins required for adaptation and survival to hypoxic stress. Although in the past two decades, the discovery of HIF-1 by Gregg Semenza et al. provided a molecular platform to investigate the mechanism underlying responses to oxygen deprivation, the molecular and cellular biology of hypoxia has still to be completely elucidated. This review summarizes recent experimental data concerned with mitochondrial structure and function adaptation to hypoxia and evaluates it in light of the main structural and functional parameters defining the mitochondrial bioenergetics. Since mitochondria contain an inhibitor protein, IF1, whose action on the F1F0 ATPase has been considered for decades of critical importance in hypoxia/ischemia, particular notice will be dedicated to analyze molecular aspects of IF1 regulation of the enzyme and its possible role in the metabolic changes induced by low oxygen levels in cells.

Mechanism(s) of HIF-1 activation

HIF-1 consists of an oxygen-sensitive HIF-1α subunit that heterodimerizes with the HIF-1β subunit to bind DNA. In high O2 tension, HIF-1α is oxidized (hydroxylated) by prolyl hydroxylases (PHDs) using α-ketoglutarate derived from the tricarboxylic acid (TCA) cycle. The hydroxylated HIF-1α subunit interacts with the von Hippel–Lindau protein, a critical member of an E3 ubiquitin ligase complex that polyubiquitylates HIF. This is then catabolized by proteasomes, such that HIF-1α is continuously synthesized and degraded under normoxic conditions [18]. Under hypoxia, HIF-1α hydroxylation does not occur, thereby stabilizing HIF-1 (Fig. 1). The active HIF-1 complex in turn binds to a core hypoxia response element in a wide array of genes involved in a diversity of biological processes, and directly transactivates glycolytic enzyme genes [29]. Notably, O2 concentration, multiple mitochondrial products, including the TCA cycle intermediates and reactive oxygen species, can coordinate PHD activity, HIF stabilization, hence the cellular responses to O2 depletion [30] and [31]. Incidentally, impaired TCA cycle flux, particularly if it is caused by succinate dehydrogenase dysfunction, results in decreased or loss of energy production from both the electron-transport chain and the Krebs cycle, and also in overproduction of free radicals [32]. This leads to severe early-onset neurodegeneration or, as it occurs in individuals carrying mutations in the non-catalytic subunits of the same enzyme, to tumors such as phaeochromocytoma and paraganglioma. However, impairment of the TCA cycle may be relevant also for the metabolic changes occurring in mitochondria exposed to hypoxia, since accumulation of succinate has been reported to inhibit PHDs [33]. It has to be noticed that some authors believe reactive oxygen species (ROS) to be essential to activate HIF-1 [34], but others challenge this idea [35], therefore the role of mitochondrial ROS in the regulation of HIF-1 under hypoxia is still controversial [36]. Moreover, the contribution of functional mitochondria to HIF-1 regulation has also been questioned by others [37][38] and [39].

http://ars.els-cdn.com/content/image/1-s2.0-S0005272810000575-gr1.jpg

Major mitochondrial changes in hypoxia

Major mitochondrial changes in hypoxia

Fig. 1. Major mitochondrial changes in hypoxia. Hypoxia could decrease electron-transport rate determining Δψm reduction, increased ROS generation, and enhanced NO synthase. One (or more) of these factors likely contributes to HIF stabilization, that in turn induces metabolic adaptation of both hypoxic cells and mitophagy. The decreased Δψm could also induce an active binding of IF1, which might change mitochondrial morphology and/or dynamics, and inhibit mitophagy. Solid lines indicate well established hypoxic changes in cells, whilst dotted lines indicate changes not yet stated. Inset, relationships between extracellular O2concentration and oxygen tension.

Oxygen is a major determinant of cell metabolism and gene expression, and as cellular O2 levels decrease, either during isolated hypoxia or ischemia-associated hypoxia, metabolism and gene expression profiles in the cells are significantly altered. Low oxygen reduces OXPHOS and Krebs cycle rates, and participates in the generation of nitric oxide (NO), which also contributes to decrease respiration rate [23] and [40]. However, oxygen is also central in the generation of reactive oxygen species, which can participate in cell signaling processes or can induce irreversible cellular damage and death [41].

As specified above, cells adapt to oxygen reduction by inducing active HIF, whose major effect on cells energy homeostasis is the inactivation of anabolism, activation of anaerobic glycolysis, and inhibition of the mitochondrial aerobic metabolism: the TCA cycle, and OXPHOS. Since OXPHOS supplies the majority of ATP required for cellular processes, low oxygen tension will severely reduce cell energy availability. This occurs through several mechanisms: first, reduced oxygen tension decreases the respiration rate, due first to nonsaturating substrate for cytochrome c oxidase (COX), secondarily, to allosteric modulation of COX[42]. As a consequence, the phosphorylation potential decreases, with enhancement of the glycolysis rate primarily due to allosteric increase of phosphofructokinase activity; glycolysis however is poorly efficient and produces lactate in proportion of 0.5 mol/mol ATP, which eventually drops cellular pH if cells are not well perfused, as it occurs under defective vasculature or ischemic conditions  [6]. Besides this “spontaneous” (thermodynamically-driven) shift from aerobic to anaerobic metabolism which is mediated by the kinetic changes of most enzymes, the HIF-1 factor activates transcription of genes encoding glucose transporters and glycolytic enzymes to further increase flux of reducing equivalents from glucose to lactate[43] and [44]. Second, HIF-1 coordinates two different actions on the mitochondrial phase of glucose oxidation: it activates transcription of the PDK1 gene encoding a kinase that phosphorylates and inactivates pyruvate dehydrogenase, thereby shunting away pyruvate from the mitochondria by preventing its oxidative decarboxylation to acetyl-CoA [45] and [46]. Moreover, HIF-1 induces a switch in the composition of cytochrome c oxidase from COX4-1 to COX4-2 isoform, which enhances the specific activity of the enzyme. As a result, both respiration rate and ATP level of hypoxic cells carrying the COX4-2 isoform of cytochrome c oxidase were found significantly increased with respect to the same cells carrying the COX4-1 isoform [47]. Incidentally, HIF-1 can also increase the expression of carbonic anhydrase 9, which catalyses the reversible hydration of CO2 to HCO3 and H+, therefore contributing to pH regulation.

Effects of hypoxia on mitochondrial structure and dynamics

Mitochondria form a highly dynamic tubular network, the morphology of which is regulated by frequent fission and fusion events. The fusion/fission machineries are modulated in response to changes in the metabolic conditions of the cell, therefore one should expect that hypoxia affect mitochondrial dynamics. Oxygen availability to cells decreases glucose oxidation, whereas oxygen shortage consumes glucose faster in an attempt to produce ATP via the less efficient anaerobic glycolysis to lactate (Pasteur effect). Under these conditions, mitochondria are not fueled with substrates (acetyl-CoA and O2), inducing major changes of structure, function, and dynamics (for a recent review see [48]). Concerning structure and dynamics, one of the first correlates that emerge is that impairment of mitochondrial fusion leads to mitochondrial depolarization, loss of mtDNA that may be accompanied by altered respiration rate, and impaired distribution of the mitochondria within cells [49][50] and [51]. Indeed, exposure of cortical neurons to moderate hypoxic conditions for several hours, significantly altered mitochondrial morphology, decreased mitochondrial size and reduced mitochondrial mean velocity. Since these effects were either prevented by exposing the neurons to inhibitors of nitric oxide synthase or mimicked by NO donors in normoxia, the involvement of an NO-mediated pathway was suggested [52]. Mitochondrial motility was also found inhibited and controlled locally by the [ADP]/[ATP] ratio [53]. Interestingly, the author used an original approach in which mitochondria were visualized using tetramethylrhodamineethylester and their movements were followed by applying single-particle tracking.

Of notice in this chapter is that enzymes controlling mitochondrial morphology regulators provide a platform through which cellular signals are transduced within the cell in order to affect mitochondrial function [54]. Accordingly, one might expect that besides other mitochondrial factors [30] and [55] playing roles in HIF stabilization, also mitochondrial morphology might reasonably be associated with HIF stabilization. In order to better define the mechanisms involved in the morphology changes of mitochondria and in their dynamics when cells experience hypoxic conditions, these pioneering studies should be corroborated by and extended to observations on other types of cells focusing also on single proteins involved in both mitochondrial fusion/fission and motion.

Effects of hypoxia on the respiratory chain complexes

O2 is the terminal acceptor of electrons from cytochrome c oxidase (Complex IV), which has a very high affinity for it, being the oxygen concentration for half-maximal respiratory rate at pH 7.4 approximately 0.7 µM [56]. Measurements of mitochondrial oxidative phosphorylation indicated that it is not dependent on oxygen concentration up to at least 20 µM at pH 7.0 and the oxygen dependence becomes markedly greater as the pH is more alkaline [56]. Similarly, Moncada et al. [57] found that the rate of O2 consumption remained constant until [O2] fell below 15 µM. Accordingly, most reports in the literature consider hypoxic conditions occurring in cells at 5–0.5% O2, a range corresponding to 46–4.6 µM O2 in the cells culture medium (see Fig. 1 inset). Since between the extracellular environment and mitochondria an oxygen pressure gradient is established [58], the O2 concentration experienced by Complex IV falls in the range affecting its kinetics, as reported above.

Under these conditions, a number of changes on the OXPHOS machinery components, mostly mediated by HIF-1 have been found. Thus, Semenza et al. [59] and others thereafter [46] reported that activation of HIF-1α induces pyruvate dehydrogenase kinase, which inhibits pyruvate dehydrogenase, suggesting that respiration is decreased by substrate limitation. Besides, other HIF-1 dependent mechanisms capable to affect respiration rate have been reported. First, the subunit composition of COX is altered in hypoxic cells by increased degradation of the COX4-1 subunit, which optimizes COX activity under aerobic conditions, and increased expression of the COX4-2 subunit, which optimizes COX activity under hypoxic conditions [29]. On the other hand, direct assay of respiration rate in cells exposed to hypoxia resulted in a significant reduction of respiration [60]. According with the evidence of Zhang et al., the respiration rate decrease has to be ascribed to mitochondrial autophagy, due to HIF-1-mediated expression of BNIP3. This interpretation is in line with preliminary results obtained in our laboratory where the assay of the citrate synthase activity of cells exposed to different oxygen tensions was performed. Fig. 2 shows the citrate synthase activity, which is taken as an index of the mitochondrial mass [11], with respect to oxygen tension: [O2] and mitochondrial mass are directly linked.

Citrate synthase activity

Citrate synthase activity

http://ars.els-cdn.com/content/image/1-s2.0-S0005272810000575-gr2.jpg

Fig. 2. Citrate synthase activity. Human primary fibroblasts, obtained from skin biopsies of 5 healthy donors, were seeded at a density of 8,000 cells/cm2 in high glucose Dulbecco’s Modified Eagle Medium, DMEM (25 mM glucose, 110 mg/l pyruvate, and 4 mM glutamine) supplemented with 15% Foetal Bovine Serum (FBS). 18 h later, cell culture dishes were washed once with Hank’s Balanced Salt Solution (HBSS) and the medium was replaced with DMEM containing 5 mM glucose, 110 mg/l pyruvate, and 4 mM glutamine supplemented with 15% FBS. Cell culture dishes were then placed into an INVIVO2 humidified hypoxia workstation (Ruskinn Technologies, Bridgend, UK) for 72 h changing the medium at 48 h, and oxygen partial pressure (tension) conditions were: 20%, 4%, 2%, 1% and 0.5%. Cells were subsequently collected within the workstation with trypsin-EDTA (0.25%), washed with PBS and resuspended in a buffer containing 10 mM Tris/HCl, 0.1 M KCl, 5 mM KH2PO4, 1 mM EGTA, 3 mM EDTA, and 2 mM MgCl2 pH 7.4 (all the solutions were preconditioned to the appropriate oxygen tension condition). The citrate synthase activity was assayed essentially by incubating 40 µg of cells with 0.02% Triton X-100, and monitoring the reaction by measuring spectrophotometrically the rate of free coenzyme A released, as described in [90]. Enzymatic activity was expressed as nmol/min/mg of protein. Three independent experiments were carried out and assays were performed in either duplicate or triplicate.

However, the observations of Semenza et al. must be seen in relation with data reported by Moncada et al.[57] and confirmed by others [61] in which it is clearly shown that when cells (various cell lines) experience hypoxic conditions, nitric oxide synthases (NOSs) are activated, therefore NO is released. As already mentioned above, NO is a strong competitor of O2 for cytochrome c oxidase, whose apparent Km results increased, hence reduction of mitochondrial cytochromes and all the other redox centres of the respiratory chain occurs. In addition, very recent data indicate a potential de-activation of Complex I when oxygen is lacking, as it occurs in prolonged hypoxia [62]. According to Hagen et al. [63] the NO-dependent inhibition of cytochrome c oxidase should allow “saved” O2 to redistribute within the cell to be used by other enzymes, including PHDs which inactivate HIF. Therefore, unless NO inhibition of cytochrome c oxidase occurs only when [O2] is very low, inhibition of mitochondrial oxygen consumption creates the paradox of a situation in which the cell may fail to register hypoxia. It has been tempted to solve this paradox, but to date only hypotheses have been proposed [23] and [26]. Interestingly, recent observations on yeast cells exposed to hypoxia revealed abnormal protein carbonylation and protein tyrosine nitration that were ascribed to increased mitochondrially generated superoxide radicals and NO, two species typically produced at low oxygen levels, that combine to form ONOO [64]. Based on these studies a possible explanation has been proposed for the above paradox.

Finally, it has to be noticed that the mitochondrial respiratory deficiency observed in cardiomyocytes of dogs in which experimental heart failure had been induced lies in the supermolecular assembly rather than in the individual components of the electron-transport chain [65]. This observation is particularly intriguing since loss of respirasomes is thought to facilitate ROS generation in mitochondria [66], therefore supercomplexes disassembly might explain the paradox of reduced [O2] and the enhanced ROS found in hypoxic cells. Specifically, hypoxia could reduce mitochondrial fusion by impairing mitochondrial membrane potential, which in turn could induce supercomplexes disassembly, increasing ROS production[11].

Complex III and ROS production

It has been estimated that, under normoxic physiological conditions, 1–2% of electron flow through the mitochondrial respiratory chain gives rise to ROS [67] and [68]. It is now recognized that the major sites of ROS production are within Complexes I and III, being prevalent the contribution of Complex I [69] (Fig. 3). It might be expected that hypoxia would decrease ROS production, due to the low level of O2 and to the diminished mitochondrial respiration [6] and [46], but ROS level is paradoxically increased. Indeed, about a decade ago, Chandel et al. [70] provided good evidence that mitochondrial reactive oxygen species trigger hypoxia-induced transcription, and a few years later the same group [71] showed that ROS generated at Complex III of the mitochondrial respiratory chain stabilize HIF-1α during hypoxia (Fig. 1 and Fig. 3). Although others have proposed mechanisms indicating a key role of mitochondria in HIF-1α regulation during hypoxia (for reviews see [64] and [72]), the contribution of mitochondria to HIF-1 regulation has been questioned by others [35][36] and [37]. Results of Gong and Agani [35] for instance show that inhibition of electron-transport Complexes I, III, and IV, as well as inhibition of mitochondrial F0F1 ATPase, prevents HIF-1α expression and that mitochondrial reactive oxygen species are not involved in HIF-1α regulation during hypoxia. Concurrently, Tuttle et al. [73], by means of a non invasive, spectroscopic approach, could find no evidence to suggest that ROS, produced by mitochondria, are needed to stabilize HIF-1α under moderate hypoxia. The same authors found the levels of HIF-1α comparable in both normal and ρ0 cells (i.e. cells lacking mitochondrial DNA). On the contrary, experiments carried out on genetic models consisting of either cells lacking cytochrome c or ρ0 cells both could evidence the essential role of mitochondrial respiration to stabilize HIF-1α [74]. Thus, cytochrome c null cells, being incapable to respire, exposed to moderate hypoxia (1.5% O2) prevented oxidation of ubiquinol and generation of the ubisemiquinone radical, thus eliminating superoxide formation at Complex III [71]. Concurrently, ρ0 cells lacking electron transport, exposed 4 h to moderate hypoxia failed to stabilize HIF-1α, suggesting the essential role of the respiratory chain for the cellular sensing of low O2 levels. In addition, recent evidence obtained on genetic manipulated cells (i.e. cytochrome b deficient cybrids) showed increased ROS levels and stabilized HIF-1α protein during hypoxia [75]. Moreover, RNA interference of the Complex III subunit Rieske iron sulfur protein in the cytochrome b deficient cells, abolished ROS generation at the Qo site of Complex III, preventing HIF-1α stabilization. These observations, substantiated by experiments with MitoQ, an efficient mitochondria-targeted antioxidant, strongly support the involvement of mitochondrial ROS in regulating HIF-1α. Nonetheless, collectively, the available data do not allow to definitely state the precise role of mitochondrial ROS in regulating HIF-1α, but the pathway stabilizing HIF-1α appears undoubtedly mitochondria-dependent [30].

Overview of mitochondrial electron and proton flux in hypoxia

Overview of mitochondrial electron and proton flux in hypoxia

Overview of mitochondrial electron and proton flux in hypoxia

http://ars.els-cdn.com/content/image/1-s2.0-S0005272810000575-gr3.jpg

Fig. 3. Overview of mitochondrial electron and proton flux in hypoxia. Electrons released from reduced cofactors (NADH and FADH2) under normoxia flow through the redox centres of the respiratory chain (r.c.) to molecular oxygen (blue dotted line), to which a proton flux from the mitochondrial matrix to the intermembrane space is coupled (blue arrows). Protons then flow back to the matrix through the F0 sector of the ATP synthase complex, driving ATP synthesis. ATP is carried to the cell cytosol by the adenine nucleotide translocator (blue arrows). Under moderate to severe hypoxia, electrons escape the r.c. redox centres and reduce molecular oxygen to the superoxide anion radical before reaching the cytochrome c (red arrow). Under these conditions, to maintain an appropriate Δψm, ATP produced by cytosolic glycolysis enters the mitochondria where it is hydrolyzed by the F1F0ATPase with extrusion of protons from the mitochondrial matrix (red arrows).

Hypoxia and ATP synthase

The F1F0 ATPase (ATP synthase) is the enzyme responsible of catalysing ADP phosphorylation as the last step of OXPHOS. It is a rotary motor using the proton motive force across the mitochondrial inner membrane to drive the synthesis of ATP [76]. It is a reversible enzyme with ATP synthesis or hydrolysis taking place in the F1 sector at the matrix side of the membrane, chemical catalysis being coupled to H+transport through the transmembrane F0 sector.

Under normoxia the enzyme synthesizes ATP, but when mitochondria experience hypoxic conditions the mitochondrial membrane potential (Δψm) decreases below its endogenous steady-state level (some 140 mV, negative inside the matrix [77]) and the F1F0 ATPase may work in the reversal mode: it hydrolyses ATP (produced by anaerobic glycolysis) and uses the energy released to pump protons from the mitochondrial matrix to the intermembrane space, concurring with the adenine nucleotide translocator (i.e. in hypoxia it exchanges cytosolic ATP4− for matrix ADP3−) to maintain the physiological Δψm ( Fig. 3). Since under conditions of limited oxygen availability the decline in cytoplasmic high energy phosphates is mainly due to hydrolysis by the ATP synthase working in reverse [6] and [78], the enzyme must be strictly regulated in order to avoid ATP dissipation. This is achieved by a natural protein, the H+ψm-dependent IF1, that binds to the catalytic F1 sector at low pH and low Δψm (such as it occurs in hypoxia/ischemia) [79]. IF1 binding to the ATP synthase results in a rapid and reversible inhibition of the enzyme [80], which could reach about 50% of maximal activity (for recent reviews see [6] and [81]).

Besides this widely studied effect, IF1 appears to be associated with ROS production and mitochondrial autophagy (mitophagy). This is a mechanism involving the catabolic degradation of macromolecules and organelles via the lysosomal pathway that contributes to housekeeping and regenerate metabolites. Autophagic degradation is involved in the regulation of the ageing process and in several human diseases, such as myocardial ischemia/reperfusion [82], Alzheimer’s Disease, Huntington diseases, and inflammatory diseases (for recent reviews see [83] and [84], and, as mentioned above, it promotes cell survival by reducing ROS and mtDNA damage under hypoxic conditions.

Campanella et al. [81] reported that, in HeLa cells under normoxic conditions, basal autophagic activity varies in relation to the expression levels of IF1. Accordingly, cells overexpressing IF1 result in ROS production similar to controls, conversely cells in which IF1 expression is suppressed show an enhanced ROS production. In parallel, the latter cells show activation of the mitophagy pathway (Fig. 1), therefore suggesting that variations in IF1 expression level may play a significant role in defining two particularly important parameters in the context of the current review: rates of ROS generation and mitophagy. Thus, the hypoxia-induced enhanced expression level of IF1[81] should be associated with a decrease of both ROS production and autophagy, which is in apparent conflict with the hypoxia-induced ROS increase and with the HIF-1-dependent mitochondrial autophagy shown by Zhang et al. [60] as an adaptive metabolic response to hypoxia. However, in the experiments of Zhang et al. the cells were exposed to hypoxia for 48 h, whereas the F1F0-ATPase inhibitor exerts a prompt action on the enzyme and to our knowledge, it has never been reported whether its action persists during prolonged hypoxic expositions. Pertinent with this problem is the very recent observation that IEX-1 (immediate early response gene X-1), a stress-inducible gene that suppresses production of ROS and protects cells from apoptosis [85], targets the mitochondrial F1F0-ATPase inhibitor for degradation, reducing ROS by decreasing Δψm. It has to be noticed that the experiments described were carried out under normal oxygen availability, but it does not seem reasonable to rule out IEX-1 from playing a role under stress conditions as those induced by hypoxia in cells, therefore this issue might deserve an investigation also at low oxygen levels.

In conclusion, data are still emerging regarding the regulation of mitochondrial function by the F1F0 ATPase within hypoxic responses in different cellular and physiological contexts. Given the broad pathophysiological role of hypoxic cellular modulation, an understanding of the subtle tuning among different effectors of the ATP synthase is desirable to eventually target future therapeutics most effectively. Our laboratory is actually involved in carrying out investigations to clarify this context.

Conclusions and perspectives

The mitochondria are important cellular platforms that both propagate and initiate intracellular signals that lead to overall cellular and metabolic responses. During the last decades, a significant amount of relevant data has been obtained on the identification of mechanisms of cellular adaptation to hypoxia. In hypoxic cells there is an enhanced transcription and synthesis of several glycolytic pathway enzymes/transporters and reduction of synthesis of proteins involved in mitochondrial catabolism. Although well defined kinetic parameters of reactions in hypoxia are lacking, it is usually assumed that these transcriptional changes lead to metabolic flux modification. The required biochemical experimentation has been scarcely addressed until now and only in few of the molecular and cellular biology studies the transporter and enzyme kinetic parameters and flux rate have been determined, leaving some uncertainties.

Central to mitochondrial function and ROS generation is an electrochemical proton gradient across the mitochondrial inner membrane that is established by the proton pumping activity of the respiratory chain, and that is strictly linked to the F1F0-ATPase function. Evaluation of the mitochondrial membrane potential in hypoxia has only been studied using semiquantitative methods based on measurements of the fluorescence intensity of probes taken up by cells experiencing normal or hypoxic conditions. However, this approach is intrinsically incorrect due to the different capability that molecular oxygen has to quench fluorescence [86] and [87] and to the uncertain concentration the probe attains within mitochondria, whose mass may be reduced by a half in hypoxia [60]. In addition, the uncertainty about measurement of mitochondrial superoxide radical and H2O2 formation in vivo [88] hampers studies on the role of mitochondrial ROS in hypoxic oxidative damage, redox signaling, and HIF-1 stabilization.

The duration and severity of hypoxic stress differentially activate the responses discussed throughout and lead to substantial phenotypic variations amongst tissues and cell models, which are not consistently and definitely known. Certainly, understanding whether a hierarchy among hypoxia response mechanisms exists and which are the precise timing and conditions of each mechanism to activate, will improve our knowledge of the biochemical mechanisms underlying hypoxia in cells, which eventually may contribute to define therapeutic targets in hypoxia-associated diseases. To this aim it might be worth investigating the hypoxia-induced structural organization of both the respiratory chain enzymes in supramolecular complexes and the assembly of the ATP synthase to form oligomers affecting ROS production [65] and inner mitochondrial membrane structure [89], respectively.

7.9.2 Hypoxia promotes isocitrate dehydrogenase-dependent carboxylation of α-ketoglutarate to citrate to support cell growth and viability

DR WisePS WardJES ShayJR CrossJJ Gruber, UM Sachdeva, et al.
Proc Nat Acad Sci Oct 27, 2011; 108(49):19611–19616
http://dx.doi.org:/10.1073/pnas.1117773108

Citrate is a critical metabolite required to support both mitochondrial bioenergetics and cytosolic macromolecular synthesis. When cells proliferate under normoxic conditions, glucose provides the acetyl-CoA that condenses with oxaloacetate to support citrate production. Tricarboxylic acid (TCA) cycle anaplerosis is maintained primarily by glutamine. Here we report that some hypoxic cells are able to maintain cell proliferation despite a profound reduction in glucose-dependent citrate production. In these hypoxic cells, glutamine becomes a major source of citrate. Glutamine-derived α-ketoglutarate is reductively carboxylated by the NADPH-linked mitochondrial isocitrate dehydrogenase (IDH2) to form isocitrate, which can then be isomerized to citrate. The increased IDH2-dependent carboxylation of glutamine-derived α-ketoglutarate in hypoxia is associated with a concomitant increased synthesis of 2-hydroxyglutarate (2HG) in cells with wild-type IDH1 and IDH2. When either starved of glutamine or rendered IDH2-deficient by RNAi, hypoxic cells are unable to proliferate. The reductive carboxylation of glutamine is part of the metabolic reprogramming associated with hypoxia-inducible factor 1 (HIF1), as constitutive activation of HIF1 recapitulates the preferential reductive metabolism of glutamine-derived α-ketoglutarate even in normoxic conditions. These data support a role for glutamine carboxylation in maintaining citrate synthesis and cell growth under hypoxic conditions.

Citrate plays a critical role at the center of cancer cell metabolism. It provides the cell with a source of carbon for fatty acid and cholesterol synthesis (1). The breakdown of citrate by ATP-citrate lyase is a primary source of acetyl-CoA for protein acetylation (2). Metabolism of cytosolic citrate by aconitase and IDH1 can also provide the cell with a source of NADPH for redox regulation and anabolic synthesis. Mammalian cells depend on the catabolism of glucose and glutamine to fuel proliferation (3). In cancer cells cultured at atmospheric oxygen tension (21% O2), glucose and glutamine have both been shown to contribute to the cellular citrate pool, with glutamine providing the major source of the four-carbon molecule oxaloacetate and glucose providing the major source of the two-carbon molecule acetyl-CoA (45). The condensation of oxaloacetate and acetyl-CoA via citrate synthase generates the 6 carbon citrate molecule. However, both the conversion of glucose-derived pyruvate to acetyl-CoA by pyruvate dehydrogenase (PDH) and the conversion of glutamine to oxaloacetate through the TCA cycle depend on NAD+, which can be compromised under hypoxic conditions. This raises the question of how cells that can proliferate in hypoxia continue to synthesize the citrate required for macromolecular synthesis.

This question is particularly important given that many cancers and stem/progenitor cells can continue proliferating in the setting of limited oxygen availability (67). Louis Pasteur first highlighted the impact of hypoxia on nutrient metabolism based on his observation that hypoxic yeast cells preferred to convert glucose into lactic acid rather than burning it in an oxidative fashion. The molecular basis for this shift in mammalian cells has been linked to the activity of the transcription factor HIF1 (810). Stabilization of the labile HIF1α subunit occurs in hypoxia. It can also occur in normoxia through several mechanisms including loss of the von Hippel-Lindau tumor suppressor (VHL), a common occurrence in renal carcinoma (11). Although hypoxia and/or HIF1α stabilization is a common feature of multiple cancers, to date the source of citrate in the setting of hypoxia or HIF activation has not been determined.

Here, we study the sources of hypoxic citrate synthesis in a glioblastoma cell line that proliferates in profound hypoxia (0.5% O2). Glucose uptake and conversion to lactic acid increased in hypoxia. However, glucose conversion into citrate dramatically declined. Glutamine consumption remained constant in hypoxia, and hypoxic cells were addicted to the use of glutamine in hypoxia as a source of α-ketoglutarate. Glutamine provided the major carbon source for citrate synthesis during hypoxia. However, the TCA cycle-dependent conversion of glutamine into citric acid was significantly suppressed. In contrast, there was a relative increase in glutamine-dependent citrate production in hypoxia that resulted from carboxylation of α-ketoglutarate. This reductive synthesis required the presence of mitochondrial isocitrate dehydrogenase 2 (IDH2). In confirmation of the reverse flux through IDH2, the increased reductive metabolism of glutamine-derived α-ketoglutarate in hypoxia was associated with increased synthesis of 2HG. Finally, constitutive HIF1α-expressing cells also demonstrated significant reductive-carboxylation-dependent synthesis of citrate in normoxia and a relative defect in the oxidative conversion of glutamine into citrate. Collectively, the data demonstrate that mitochondrial glutamine metabolism can be rerouted through IDH2-dependent citrate synthesis in support of hypoxic cell growth.

Some Cancer Cells Can Proliferate at 0.5% O2 Despite a Sharp Decline in Glucose-Dependent Citrate Synthesis.

At 21% O2, cancer cells have been shown to synthesize citrate by condensing glucose-derived acetyl-CoA with glutamine-derived oxaloacetate through the activity of the canonical TCA cycle enzyme citrate synthase (4). In contrast, less is known regarding the synthesis of citrate by cells that can continue proliferating in hypoxia. The glioblastoma cell line SF188 is able to proliferate at 0.5% O2 (Fig. 1A), a level of hypoxia that is sufficient to stabilize HIF1α (Fig. 1B) and predicted to limit respiration (1213). Consistent with previous observations in hypoxic cells, we found that SF188 cells demonstrated increased lactate production when incubated in hypoxia (Fig. 1C), and the ratio of lactate produced to glucose consumed increased demonstrating an increase in the rate of anaerobic glycolysis. When glucose-derived carbon in the form of pyruvate is converted to lactate, it is diverted away from subsequent metabolism that can contribute to citrate production. However, we observed that SF188 cells incubated in hypoxia maintain their intracellular citrate to ∼75% of the level maintained under normoxia (Fig. 1D). This prompted an investigation of how proliferating cells maintain citrate production under hypoxia.

SF188 glioblastoma cells proliferate at 0.5% O2 despite a profound reduction in glucose-dependent citrate synthesis.

SF188 glioblastoma cells proliferate at 0.5% O2 despite a profound reduction in glucose-dependent citrate synthesis.

http://www.pnas.org/content/108/49/19611/F1.medium.gif

Fig. 1. SF188 glioblastoma cells proliferate at 0.5% O2 despite a profound reduction in glucose-dependent citrate synthesis. (A) SF188 cells were plated in complete medium equilibrated with 21% O2 (Normoxia) or 0.5% O2 (Hypoxia), total viable cells were counted 24 h and 48 h later (Day 1 and Day 2), and population doublings were calculated. Data are the mean ± SEM of four independent experiments. (B) Western blot demonstrates stabilized HIF1α protein in cells cultured in hypoxia compared with normoxia. (C) Cells were grown in normoxia or hypoxia for 24 h, after which culture medium was collected. Medium glucose and lactate levels were measured and compared with the levels in fresh medium. (D) Cells were cultured for 24 h as in C. Intracellular metabolism was then quenched with 80% MeOH prechilled to −80 °C that was spiked with a 13C-labeled citrate as an internal standard. Metabolites were then extracted, and intracellular citrate levels were analyzed with GC-MS and normalized to cell number. Data for C and D are the mean ± SEM of three independent experiments. (E) Model depicting the pathway for cit+2 production from [U-13C]glucose. Glucose uniformly 13C-labeled will generate pyruvate+3. Pyruvate+3 can be oxidatively decarboxylated by PDH to produce acetyl-CoA+2, which can condense with unlabeled oxaloacetate to produce cit+2. (F) Cells were cultured for 24 h as in C and D, followed by an additional 4 h of culture in glucose-deficient medium supplemented with 10 mM [U-13C]glucose. Intracellular metabolites were then extracted, and 13C-enrichment in cellular citrate was analyzed by GC-MS and normalized to the total citrate pool size. Data are the mean ± SD of three independent cultures from a representative of two independent experiments. *P < 0.05, ***P < 0.001.

Increased glucose uptake and glycolytic metabolism are critical elements of the metabolic response to hypoxia. To evaluate the contributions made by glucose to the citrate pool under normoxia or hypoxia, SF188 cells incubated in normoxia or hypoxia were cultured in medium containing 10 mM [U-13C]glucose. Following a 4-h labeling period, cellular metabolites were extracted and analyzed for isotopic enrichment by gas chromatography-mass spectrometry (GC-MS). In normoxia, the major 13C-enriched citrate species found was citrate enriched with two 13C atoms (cit+2), which can arise from the NAD+-dependent decarboxylation of pyruvate+3 to acetyl-CoA+2 by PDH, followed by the condensation of acetyl-CoA+2 with unenriched oxaloacetate (Fig. 1 E and F). Compared with the accumulation of cit+2, we observed minimal accumulation of cit+3 and cit+5 under normoxia. Cit+3 arises from pyruvate carboxylase (PC)-dependent conversion of pyruvate+3 to oxaloacetate+3, followed by the condensation of oxaloacetate+3 with unenriched acetyl-CoA. Cit+5 arises when PC-generated oxaloacetate+3 condenses with PDH-generated acetyl-CoA+2. The lack of cit+3 and cit+5 accumulation is consistent with PC activity not playing a major role in citrate production in normoxic SF188 cells, as reported (4).

In hypoxic cells, the major citrate species observed was unenriched. Cit+2, cit+3, and cit+5 all constituted minor fractions of the total citrate pool, consistent with glucose carbon not being incorporated into citrate through either PDH or PC-mediated metabolism under hypoxic conditions (Fig. 1F). These data demonstrate that in contrast to normoxic cells, where a large percentage of citrate production depends on glucose-derived carbon, hypoxic cells significantly reduce their rate of citrate production from glucose.

Glutamine Carbon Metabolism Is Required for Viability in Hypoxia.

In addition to glucose, we have previously reported that glutamine can contribute to citrate production during cell growth under normoxic conditions (4). Surprisingly, under hypoxic conditions, we observed that SF188 cells retained their high rate of glutamine consumption (Fig. 2A). Moreover, hypoxic cells cultured in glutamine-deficient medium displayed a significant loss of viability (Fig. 2B). In normoxia, the requirement for glutamine to maintain viability of SF188 cells can be satisfied by α-ketoglutarate, the downstream metabolite of glutamine that is devoid of nitrogenous groups (14). α-ketoglutarate cannot fulfill glutamine’s roles as a nitrogen source for nonessential amino acid synthesis or as an amide donor for nucleotide or hexosamine synthesis, but can be metabolized through the oxidative TCA cycle to regenerate oxaloacetate, and subsequently condense with glucose-derived acetyl-CoA to produce citrate. To test whether the restoration of carbon from glutamine metabolism in the form of α-ketoglutarate could rescue the viability defect of glutamine-starved SF188 cells even under hypoxia, SF188 cells incubated in hypoxia were cultured in glutamine-deficient medium supplemented with a cell-penetrant form of α-ketoglutarate (dimethyl α-ketoglutarate). The addition of dimethyl α-ketoglutarate rescued the defect in cell viability observed upon glutamine withdrawal (Fig. 2B). These data demonstrate that, even under hypoxic conditions, when the ability of glutamine to replenish oxaloacetate through oxidative TCA cycle metabolism is diminished, SF188 cells retain their requirement for glutamine as the carbon backbone for α-ketoglutarate. This result raised the possibility that glutamine could be the carbon source for citrate production through an alternative, nonoxidative, pathway in hypoxia.

Glutamine carbon is required for hypoxic cell viability

Glutamine carbon is required for hypoxic cell viability

Glutamine carbon is required for hypoxic cell viability

http://www.pnas.org/content/108/49/19611/F2.medium.gif

Fig. 2. Glutamine carbon is required for hypoxic cell viability and contributes to increased citrate production through reductive carboxylation relative to oxidative metabolism in hypoxia. (A) SF188 cells were cultured for 24 h in complete medium equilibrated with either 21% O2 (Normoxia) or 0.5% O2(Hypoxia). Culture medium was then removed from cells and analyzed for glutamine levels which were compared with the glutamine levels in fresh medium. Data are the mean ± SEM of three independent experiments. (B) The requirement for glutamine to maintain hypoxic cell viability can be satisfied by α-ketoglutarate. Cells were cultured in complete medium equilibrated with 0.5% O2 for 24 h, followed by an additional 48 h at 0.5% O2 in either complete medium (+Gln), glutamine-deficient medium (−Gln), or glutamine-deficient medium supplemented with 7 mM dimethyl α-ketoglutarate (−Gln +αKG). All medium was preconditioned in 0.5% O2. Cell viability was determined by trypan blue dye exclusion. Data are the mean and range from two independent experiments. (C) Model depicting the pathways for cit+4 and cit+5 production from [U-13C]glutamine (glutamine+5). Glutamine+5 is catabolized to α-ketoglutarate+5, which can then contribute to citrate production by two divergent pathways. Oxidative metabolism produces oxaloacetate+4, which can condense with unlabeled acetyl-CoA to produce cit+4. Alternatively, reductive carboxylation produces isocitrate+5, which can isomerize to cit+5. (D) Glutamine contributes to citrate production through increased reductive carboxylation relative to oxidative metabolism in hypoxic proliferating cancer cells. Cells were cultured for 24 h as in A, followed by 4 h of culture in glutamine-deficient medium supplemented with 4 mM [U-13C]glutamine. 13C enrichment in cellular citrate was quantitated with GC-MS. Data are the mean ± SD of three independent cultures from a representative of three independent experiments. **P < 0.01.

Cells Proliferating in Hypoxia Maintain Levels of Additional Metabolites Through Reductive Carboxylation.

Previous work has documented that, in normoxic conditions, SF188 cells use glutamine as the primary anaplerotic substrate, maintaining the pool sizes of TCA cycle intermediates through oxidative metabolism (4). Surprisingly, we found that, when incubated in hypoxia, SF188 cells largely maintained their levels of aspartate (in equilibrium with oxaloacetate), malate, and fumarate (Fig. 3A). To distinguish how glutamine carbon contributes to these metabolites in normoxia and hypoxia, SF188 cells incubated in normoxia or hypoxia were cultured in medium containing 4 mM [U-13C]glutamine. After a 4-h labeling period, metabolites were extracted and the intracellular pools of aspartate, malate, and fumarate were analyzed by GC-MS.

In normoxia, the majority of the enriched intracellular asparatate, malate, and fumarate were the +4 species, which arise through oxidative metabolism of glutamine-derived α-ketoglutarate (Fig. 3 B and C). The +3 species, which can be derived from the citrate generated by the reductive carboxylation of glutamine-derived α-ketoglutarate, constituted a significantly lower percentage of the total aspartate, malate, and fumarate pools. By contrast, in hypoxia, the +3 species constituted a larger percentage of the total aspartate, malate, and fumarate pools than they did in normoxia. These data demonstrate that, in addition to citrate, hypoxic cells preferentially synthesize oxaloacetate, malate, and fumarate through the pathway of reductive carboxylation rather than the oxidative TCA cycle.

IDH2 Is Critical in Hypoxia for Reductive Metabolism of Glutamine and for Cell Proliferation.

We hypothesized that the relative increase in reductive carboxylation we observed in hypoxia could arise from the suppression of α-ketoglutarate oxidation through the TCA cycle. Consistent with this, we found that α-ketoglutarate levels increased in SF188 cells following 24 h in hypoxia (Fig. 4A). Surprisingly, we also found that levels of the closely related metabolite 2-hydroxyglutarate (2HG) increased in hypoxia, concomitant with the increase in α-ketoglutarate under these conditions. 2HG can arise from the noncarboxylating reduction of α-ketoglutarate (Fig. 4B). Recent work has found that specific cancer-associated mutations in the active sites of either IDH1 or IDH2 lead to a 10- to 100-fold enhancement in this activity facilitating 2HG production (1517), but SF188 cells lack IDH1/2 mutations. However, 2HG levels are also substantially elevated in the inborn error of metabolism 2HG aciduria, and the majority of patients with this disease lack IDH1/2 mutations. As 2HG has been demonstrated to arise in these patients from mitochondrial α-ketoglutarate (18), we hypothesized that both the increased reductive carboxylation of glutamine-derived α-ketoglutarate to citrate and the increased 2HG accumulation we observed in hypoxia could arise from increased reductive metabolism by wild-type IDH2 in the mitochondria.

Reductive carboxylation of glutamine-derived α-ketoglutarate to citrate in hypoxic cancer cells is dependent on mitochondrial IDH2

Reductive carboxylation of glutamine-derived α-ketoglutarate to citrate in hypoxic cancer cells is dependent on mitochondrial IDH2

Reductive carboxylation of glutamine-derived α-ketoglutarate to citrate in hypoxic cancer cells is dependent on mitochondrial IDH2

http://www.pnas.org/content/108/49/19611/F4.medium.gif

Fig. 4. Reductive carboxylation of glutamine-derived α-ketoglutarate to citrate in hypoxic cancer cells is dependent on mitochondrial IDH2. (A) α-ketoglutarate and 2HG increase in hypoxia. SF188 cells were cultured in complete medium equilibrated with either 21% O2 (Normoxia) or 0.5% O2 (Hypoxia) for 24 h. Intracellular metabolites were then extracted, cell extracts spiked with a 13C-labeled citrate as an internal standard, and intracellular α-ketoglutarate and 2HG levels were analyzed with GC-MS. Data shown are the mean ± SEM of three independent experiments. (B) Model for reductive metabolism from glutamine-derived α-ketoglutarate. Glutamine+5 is catabolized to α-ketoglutarate+5. Carboxylation of α-ketoglutarate+5 followed by reduction of the carboxylated intermediate (reductive carboxylation) will produce isocitrate+5, which can then isomerize to cit+5. In contrast, reductive activity on α-ketoglutarate+5 that is uncoupled from carboxylation will produce 2HG+5. (C) IDH2 is required for reductive metabolism of glutamine-derived α-ketoglutarate in hypoxia. SF188 cells transfected with a siRNA against IDH2 (siIDH2) or nontargeting negative control (siCTRL) were cultured for 2 d in complete medium equilibrated with 0.5% O2. (Upper) Cells were then cultured at 0.5% O2 for an additional 4 h in glutamine-deficient medium supplemented with 4 mM [U-13C]glutamine. 13C enrichment in intracellular citrate and 2HG was determined and normalized to the relevant metabolite total pool size. (Lower) Cells transfected and cultured in parallel at 0.5% O2 were counted by hemacytometer (excluding nonviable cells with trypan blue staining) or harvested for protein to assess IDH2 expression by Western blot. Data shown for GC-MS and cell counts are the mean ± SD of three independent cultures from a representative experiment. **P < 0.01, ***P < 0.001.

In an experiment to test this hypothesis, SF188 cells were transfected with either siRNA directed against mitochondrial IDH2 (siIDH2) or nontargeting control, incubated in hypoxia for 2 d, and then cultured for another 4 h in hypoxia in media containing 4 mM [U-13C]glutamine. After the labeling period, metabolites were extracted and analyzed by GC-MS (Fig. 4C). Hypoxic SF188 cells transfected with siIDH2 displayed a decreased contribution of cit+5 to the total citrate pool, supporting an important role for IDH2 in the reductive carboxylation of glutamine-derived α-ketoglutarate in hypoxic conditions. The contribution of cit+4 to the total citrate pool did not decrease with siIDH2 treatment, consistent with IDH2 knockdown specifically affecting the pathway of reductive carboxylation and not other fundamental TCA cycle-regulating processes. In confirmation of reverse flux occurring through IDH2, the contribution of 2HG+5 to the total 2HG pool decreased in siIDH2-treated cells. Supporting the importance of citrate production by IDH2-mediated reductive carboxylation for hypoxic cell proliferation, siIDH2-transfected SF188 cells displayed a defect in cellular accumulation in hypoxia. Decreased expression of IDH2 protein following siIDH2 transfection was confirmed by Western blot. Collectively, these data point to the importance of mitochondrial IDH2 for the increase in reductive carboxylation flux of glutamine-derived α-ketoglutarate to maintain citrate levels in hypoxia, and to the importance of this reductive pathway for hypoxic cell proliferation.

Reprogramming of Metabolism by HIF1 in the Absence of Hypoxia Is Sufficient to Induce Increased Citrate Synthesis by Reductive Carboxylation Relative to Oxidative Metabolism.

The relative increase in the reductive metabolism of glutamine-derived α-ketoglutarate at 0.5% O2 may be explained by the decreased ability to carry out oxidative NAD+-dependent reactions as respiration is inhibited (1213). However, a shift to preferential reductive glutamine metabolism could also result from the active reprogramming of cellular metabolism by HIF1 (810), which inhibits the generation of mitochondrial acetyl-CoA necessary for the synthesis of citrate by oxidative glucose and glutamine metabolism (Fig. 5A). To better understand the role of HIF1 in reductive glutamine metabolism, we used VHL-deficient RCC4 cells, which display constitutive expression of HIF1α under normoxia (Fig. 5B). RCC4 cells expressing either a nontargeting control shRNA (shCTRL) or an shRNA directed at HIF1α (shHIF1α) were incubated in normoxia and cultured in medium with 4 mM [U-13C]glutamine. Following a 4-h labeling period, metabolites were extracted and the cellular citrate pool was analyzed by GC-MS. In shCTRL cells, which have constitutive HIF1α expression despite incubation in normoxia, the majority of the total citrate pool was constituted by the cit+5 species, with low levels of all other species including cit+4 (Fig. 5C). By contrast, in HIF1α-deficient cells the contribution of cit+5 to the total citrate pool was greatly decreased, whereas the contribution of cit+4 to the total citrate pool increased and was the most abundant citrate species. These data demonstrate that the relative enhancement of the reductive carboxylation pathway for citrate synthesis can be recapitulated by constitutive HIF1 activation in normoxia.

Reprogramming of metabolism by HIF1 in the absence of hypoxia

Reprogramming of metabolism by HIF1 in the absence of hypoxia

http://www.pnas.org/content/108/49/19611/F5.medium.gif

Reprogramming of metabolism by HIF1 in the absence of hypoxia is sufficient to induce reductive carboxylation of glutamine-derived α-ketoglutarate.

Fig. 5. Reprogramming of metabolism by HIF1 in the absence of hypoxia is sufficient to induce reductive carboxylation of glutamine-derived α-ketoglutarate. (A) Model depicting how HIF1 signaling’s inhibition of pyruvate dehydrogenase (PDH) activity and promotion of lactate dehydrogenase-A (LDH-A) activity can block the generation of mitochondrial acetyl-CoA from glucose-derived pyruvate, thereby favoring citrate synthesis from reductive carboxylation of glutamine-derived α-ketoglutarate. (B) Western blot demonstrating HIF1α protein in RCC4 VHL−/− cells in normoxia with a nontargeting shRNA (shCTRL), and the decrease in HIF1α protein in RCC4 VHL−/− cells stably expressing HIF1α shRNA (shHIF1α). (C) HIF1-induced reprogramming of glutamine metabolism. Cells from B at 21% O2 were cultured for 4 h in glutamine-deficient medium supplemented with 4 mM [U-13C]glutamine. Intracellular metabolites were then extracted, and 13C enrichment in cellular citrate was determined by GC-MS. Data shown are the mean ± SD of three independent cultures from a representative of three independent experiments. ***P < 0.001.

Compared with glucose metabolism, much less is known regarding how glutamine metabolism is altered under hypoxia. It has also remained unclear how hypoxic cells can maintain the citrate production necessary for macromolecular biosynthesis. In this report, we demonstrate that in contrast to cells at 21% O2, where citrate is predominantly synthesized through oxidative metabolism of both glucose and glutamine, reductive carboxylation of glutamine carbon becomes the major pathway of citrate synthesis in cells that can effectively proliferate at 0.5% O2. Moreover, we show that in these hypoxic cells, reductive carboxylation of glutamine-derived α-ketoglutarate is dependent on mitochondrial IDH2. Although others have previously suggested the existence of reductive carboxylation in cancer cells (1920), these studies failed to demonstrate the intracellular localization or specific IDH isoform responsible for the reductive carboxylation flux. Recently, we identified IDH2 as an isoform that contributes to reductive carboxylation in cancer cells incubated at 21% O2 (16), but remaining unclear were the physiological importance and regulation of this pathway relative to oxidative metabolism, as well as the conditions where this reductive pathway might be advantageous for proliferating cells.

Here we report that IDH2-mediated reductive carboxylation of glutamine-derived α-ketoglutarate to citrate is an important feature of cells proliferating in hypoxia. Moreover, the reliance on reductive glutamine metabolism can be recapitulated in normoxia by constitutive HIF1 activation in cells with loss of VHL. The mitochondrial NADPH/NADP+ ratio required to fuel the reductive reaction through IDH2 can arise from the increased NADH/NAD+ ratio existing in the mitochondria under hypoxic conditions (2122), with the transfer of electrons from NADH to NADP+ to generate NADPH occurring through the activity of the mitochondrial transhydrogenase (23). Our data do not exclude a complementary role for cytosolic IDH1 in impacting reductive glutamine metabolism, potentially through its oxidative function in an IDH2/IDH1 shuttle that transfers high energy electrons in the form of NADPH from mitochondria to cytosol (1624).

In further support of the increased mitochondrial reductive glutamine metabolism that we observe in hypoxia, we report here that incubation in hypoxia can lead to elevated 2HG levels in cells lacking IDH1/2 mutations. 2HG production from glutamine-derived α-ketoglutarate significantly decreased with knockdown of IDH2, supporting the conclusion that 2HG is produced in hypoxia by enhanced reverse flux of α-ketoglutarate through IDH2 in a truncated, noncarboxylating reductive reaction. However, other mechanisms may also contribute to 2HG elevation in hypoxia. These include diminished oxidative activity and/or enhanced reductive activity of the 2HG dehydrogenase, a mitochondrial enzyme that normally functions to oxidize 2HG back to α-ketoglutarate (25). The level of 2HG elevation we observe in hypoxic cells is associated with a concomitant increase in α-ketoglutarate, and is modest relative to that observed in cancers with IDH1/2 gain-of-function mutations. Nonetheless, 2HG elevation resulting from hypoxia in cells with wild-type IDH1/2 may hold promise as a cellular or serum biomarker for tissues undergoing chronic hypoxia and/or excessive glutamine metabolism.

The IDH2-dependent reductive carboxylation pathway that we propose in this report allows for continued citrate production from glutamine carbon when hypoxia and/or HIF1 activation prevents glucose carbon from contributing to citrate synthesis. Moreover, as opposed to continued oxidative TCA cycle functioning in hypoxia which can increase reactive oxygen species (ROS), reductive carboxylation of α-ketoglutarate in the mitochondria may serve as an electron sink that decreases the generation of ROS. HIF1 activity is not limited to the setting of hypoxia, as a common feature of several cancers is the normoxic stabilization of HIF1α through loss of the VHL tumor suppressor or other mechanisms. We demonstrate here that altered glutamine metabolism through a mitochondrial reductive pathway is a central aspect of hypoxic proliferating cell metabolism and HIF1-induced metabolic reprogramming. These findings are relevant for the understanding of numerous constitutive HIF1-expressing malignancies, as well as for populations, such as stem progenitor cells, which frequently proliferate in hypoxic conditions.

7.9.3 Hypoxia-Inducible Factors in Physiology and Medicine

Gregg L. Semenza
Cell. 2012 Feb 3; 148(3): 399–408.
http://dx.doi.org/10.1016%2Fj.cell.2012.01.021

Oxygen homeostasis represents an organizing principle for understanding metazoan evolution, development, physiology, and pathobiology. The hypoxia-inducible factors (HIFs) are transcriptional activators that function as master regulators of oxygen homeostasis in all metazoan species. Rapid progress is being made in elucidating homeostatic roles of HIFs in many physiological systems, determining pathological consequences of HIF dysregulation in chronic diseases, and investigating potential targeting of HIFs for therapeutic purposes. Oxygen homeostasis represents an organizing principle for understanding metazoan evolution, development, physiology, and pathobiology. The hypoxia-inducible factors (HIFs) are transcriptional activators that function as master regulators of oxygen homeostasis in all metazoan species. Rapid progress is being made in elucidating homeostatic roles of HIFs in many physiological systems, determining pathological consequences of HIF dysregulation in chronic diseases, and investigating potential targeting of HIFs for therapeutic purposes.

 

Oxygen is central to biology because of its utilization in the process of respiration. O2 serves as the final electron acceptor in oxidative phosphorylation, which carries with it the risk of generating reactive oxygen species (ROS) that react with cellular macromolecules and alter their biochemical or physical properties, resulting in cell dysfunction or death. As a consequence, metazoan organisms have evolved elaborate cellular metabolic and systemic physiological systems that are designed to maintain oxygen homeostasis. This review will focus on the role of hypoxia-inducible factors (HIFs) as master regulators of oxygen homeostasis and, in particular, on recent advances in understanding their roles in physiology and medicine. Due to space limitations and the remarkably pleiotropic effects of HIFs, the description of such roles will be illustrative rather than comprehensive.

O2 and Evolution, Part 1

Accumulation of O2 in Earth’s atmosphere starting ~2.5 billion years ago led to evolution of the extraordinarily efficient system of oxidative phosphorylation that transfers chemical energy stored in carbon bonds of organic molecules to the high-energy phosphate bond in ATP, which is used to power physicochemical reactions in living cells. Energy produced by mitochondrial respiration is sufficient to power the development and maintenance of multicellular organisms, which could not be sustained by energy produced by glycolysis alone (Lane and Martin, 2010). The modest dimensions of primitive metazoan species were such that O2 could diffuse from the atmosphere to all of the organism’s thousand cells, as is the case for the worm Caenorhabditis elegans. To escape the constraints placed on organismal growth by diffusion, systems designed to conduct air to cells deep within the body evolved and were sufficient for O2delivery to organisms with hundreds of thousands of cells, such as the fly Drosophila melanogaster. The final leap in body scale occurred in vertebrates and was associated with the evolution of complex respiratory, circulatory, and nervous systems designed to efficiently capture and distribute O2 to hundreds of millions of millions of cells in the case of the adult Homo sapiens.

Hypoxia-Inducible Factors

Hypoxia-inducible factor 1 (HIF-1) is expressed by all extant metazoan species analyzed (Loenarz et al., 2011). HIF-1 consists of HIF-1α and HIF-1β subunits, which each contain basic helix-loop-helix-PAS (bHLH-PAS) domains (Wang et al., 1995) that mediate heterodimerization and DNA binding (Jiang et al., 1996a). HIF-1β heterodimerizes with other bHLH-PAS proteins and is present in excess, such that HIF-1α protein levels determine HIF-1 transcriptional activity (Semenza et al., 1996).

Under well-oxygenated conditions, HIF-1α is bound by the von Hippel-Lindau (VHL) protein, which recruits an ubiquitin ligase that targets HIF-1α for proteasomal degradation (Kaelin and Ratcliffe, 2008). VHL binding is dependent upon hydroxylation of a specific proline residue in HIF-1α by the prolyl hydroxylase PHD2, which uses O2 as a substrate such that its activity is inhibited under hypoxic conditions (Epstein et al., 2001). In the reaction, one oxygen atom is inserted into the prolyl residue and the other atom is inserted into the co-substrate α-ketoglutarate, splitting it into CO2 and succinate (Kaelin and Ratcliffe, 2008). Factor inhibiting HIF-1 (FIH-1) represses HIF-1α transactivation function (Mahon et al., 2001) by hydroxylating an asparaginyl residue, using O2 and α-ketoglutarate as substrates, thereby blocking the association of HIF-1α with the p300 coactivator protein (Lando et al., 2002). Dimethyloxalylglycine (DMOG), a competitive antagonist of α-ketoglutarate, inhibits the hydroxylases and induces HIF-1-dependent transcription (Epstein et al., 2001). HIF-1 activity is also induced by iron chelators (such as desferrioxamine) and cobalt chloride, which inhibit hydroxylases by displacing Fe(II) from the catalytic center (Epstein et al., 2001).

Studies in cultured cells (Jiang et al., 1996b) and isolated, perfused, and ventilated lung preparations (Yu et al., 1998) revealed an exponential increase in HIF-1α levels at O2 concentrations less than 6% (~40 mm Hg), which is not explained by known biochemical properties of the hydroxylases. In most adult tissues, O2concentrations are in the range of 3-5% and any decrease occurs along the steep portion of the dose-response curve, allowing a graded response to hypoxia. Analyses of cultured human cells have revealed that expression of hundreds of genes was increased in response to hypoxia in a HIF-1-dependent manner (as determined by RNA interference) with direct binding of HIF-1 to the gene (as determined by chromatin immunoprecipitation [ChIP] assays); in addition, the expression of hundreds of genes was decreased in response to hypoxia in a HIF-1-dependent manner but binding of HIF-1 to these genes was not detected (Mole et al., 2009), indicating that HIF-dependent repression occurs via indirect mechanisms, which include HIF-1-dependent expression of transcriptional repressors (Yun et al., 2002) and microRNAs (Kulshreshtha et al., 2007). ChIP-seq studies have revealed that only 40% of HIF-1 binding sites are located within 2.5 kb of the transcription start site (Schödel et al., 2011).

In vertebrates, HIF-2α is a HIF-1α paralog that is also regulated by prolyl and asparaginyl hydroxylation and dimerizes with HIF-1β, but is expressed in a cell-restricted manner and plays important roles in erythropoiesis, vascularization, and pulmonary development, as described below. In D. melanogaster, the gene encoding the HIF-1α ortholog is designated similar and its paralog is designated trachealess because inactivating mutations result in defective development of the tracheal tubes (Wilk et al., 1996). In contrast, C. elegans has only a single HIF-1α homolog (Epstein et al., 2001). Thus, in both invertebrates and vertebrates, evolution of specialized systems for O2 delivery was associated with the appearance of a HIF-1α paralog.

O2 and Metabolism

The regulation of metabolism is a principal and primordial function of HIF-1. Under hypoxic conditions, HIF-1 mediates a transition from oxidative to glycolytic metabolism through its regulation of: PDK1, encoding pyruvate dehydrogenase (PDH) kinase 1, which phosphorylates and inactivates PDH, thereby inhibiting the conversion of pyruvate to acetyl coenzyme A for entry into the tricarboxylic acid cycle (Kim et al., 2006Papandreou et al., 2006); LDHA, encoding lactate dehydrogenase A, which converts pyruvate to lactate (Semenza et al. 1996); and BNIP3 (Zhang et al. 2008) and BNIP3L (Bellot et al., 2009), which mediate selective mitochondrial autophagy (Figure 1). HIF-1 also mediates a subunit switch in cytochrome coxidase that improves the efficiency of electron transfer under hypoxic conditions (Fukuda et al., 2007). An analogous subunit switch is also observed in Saccharomyces cerevisiae, although it is mediated by a completely different mechanism (yeast lack HIF-1), suggesting that it may represent a fundamental response of eukaryotic cells to hypoxia.

Regulation of Glucose Metabolism nihms-350382-f0001

Regulation of Glucose Metabolism nihms-350382-f0001

Regulation of Glucose Metabolism

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3437543/bin/nihms-350382-f0001.gif
Figure 1
Regulation of Glucose Metabolism

It is conventional wisdom that cells switch to glycolysis when O2 becomes limiting for mitochondrial ATP production. Yet, HIF-1α-null mouse embryo fibroblasts, which do not down-regulate respiration under hypoxic conditions, have higher ATP levels at 1% O2 than wild-type cells at 20% O2, demonstrating that under these conditions O2 is not limiting for ATP production (Zhang et al., 2008). However, the HIF-1α-null cells die under prolonged hypoxic conditions due to ROS toxicity (Kim et al. 2006Zhang et al., 2008). These studies have led to a paradigm shift with regard to our understanding of the regulation of cellular metabolism (Semenza, 2011): the purpose of this switch is to prevent excess mitochondrial generation of ROS that would otherwise occur due to the reduced efficiency of electron transfer under hypoxic conditions (Chandel et al., 1998). This may be particularly important in stem cells, in which avoidance of DNA damage is critical (Suda et al., 2011).

Role of HIFs in Development

Much of mammalian embryogenesis occurs at O2 concentrations of 1-5% and O2 functions as a morphogen (through HIFs) in many developmental systems (Dunwoodie, 2009). Mice that are homozygous for a null allele at the locus encoding HIF-1α die by embryonic day 10.5 with cardiac malformations, vascular defects, and impaired erythropoiesis, indicating that all three components of the circulatory system are dependent upon HIF-1 for normal development (Iyer et al., 1998Yoon et al., 2011). Depending on the genetic background, mice lacking HIF-2α: die by embryonic day 12.5 with vascular defects (Peng et al., 2000) or bradycardia due to deficient catecholamine production (Tian et al., 1998); die as neonates due to impaired lung maturation (Compernolle et al., 2002); or die several months after birth due to ROS-mediated multi-organ failure (Scortegagna et al., 2003). Thus, while vertebrate evolution was associated with concomitant appearance of the circulatory system and HIF-2α, both HIF-1 and HIF-2 have important roles in circulatory system development. Conditional knockout of HIF-1α in specific cell types has demonstrated important roles in chondrogenesis (Schipani et al., 2001), adipogenesis (Yun et al., 2002), B-lymphocyte development (Kojima et al., 2002), osteogenesis (Wang et al., 2007), hematopoiesis (Takubo et al., 2010), T-lymphocyte differentiation (Dang et al., 2011), and innate immunity (Zinkernagel et al., 2007). While knockout mouse experiments point to the adverse effects of HIF-1 loss-of-function on development, it is also possible that increased HIF-1 activity, induced by hypoxia in embryonic tissues as a result of abnormalities in placental blood flow, may also dysregulate development and result in congenital malformations. For example, HIF-1α has been shown to interact with, and stimulate the transcriptional activity of, Notch, which plays a key role in many developmental pathways (Gustafsson et al., 2005).

Translational Prospects

Drug discovery programs have been initiated at many pharmaceutical and biotech companies to develop prolyl hydroxylase inhibitors (PHIs) that, as described above for DMOG, induce HIF activity for treatment of disorders in which HIF mediates protective physiological responses. Local and/or short term induction of HIF activity by PHIs, gene therapy, or other means are likely to be useful novel therapies for many of the diseases described above. In the case of ischemic cardiovascular disease, local therapy is needed to provide homing signals for the recruitment of BMDACs. Chronic systemic use of PHIs must be approached with great caution: individuals with genetic mutations that constitutively activate the HIF pathway (described below) have increased incidence of cardiovascular disease and mortality (Yoon et al., 2011). On the other hand, the profound inhibition of HIF activity and vascular responses to ischemia that are associated with aging suggest that systemic replacement therapy might be contemplated as a preventive measure for subjects in whom impaired HIF responses to hypoxia can be documented. In C. elegans, VHL loss-of-function increases lifespan in a HIF-1-dependent manner (Mehta et al., 2009), providing further evidence for a mutually antagonistic relationship between HIF-1 and aging.

Cancer

Cancers contain hypoxic regions as a result of high rates of cell proliferation coupled with the formation of vasculature that is structurally and functionally abnormal. Increased HIF-1α and/or HIF-2α levels in diagnostic tumor biopsies are associated with increased risk of mortality in cancers of the bladder, brain, breast, colon, cervix, endometrium, head/neck, lung, ovary, pancreas, prostate, rectum, and stomach; these results are complemented by experimental studies, which demonstrate that genetic manipulations that increase HIF-1α expression result in increased tumor growth, whereas loss of HIF activity results in decreased tumor growth (Semenza, 2010). HIFs are also activated by genetic alterations, most notably, VHL loss of function in clear cell renal carcinoma (Majmunder et al., 2010). HIFs activate transcription of genes that play key roles in critical aspects of cancer biology, including stem cell maintenance (Wang et al., 2011), cell immortalization, epithelial-mesenchymal transition (Mak et al., 2010), genetic instability (Huang et al., 2007), vascularization (Liao and Johnson, 2007), glucose metabolism (Luo et al., 2011), pH regulation (Swietach et al., 2007), immune evasion (Lukashev et al., 2007), invasion and metastasis (Chan and Giaccia, 2007), and radiation resistance (Moeller et al., 2007). Given the extensive validation of HIF-1 as a potential therapeutic target, drugs that inhibit HIF-1 have been identified and shown to have anti-cancer effects in xenograft models (Table 1Semenza, 2010).

Table 1  Drugs that Inhibit HIF-1

Process Inhibited Drug Class Prototype
HIF-1 α synthesis Cardiac glycosidemTOR inhibitorMicrotubule targeting agent

Topoisomerase I inhibitor

DigoxinRapamycin2-Methoxyestradiol

Topotecan

HIF-1 α protein stability HDAC inhibitorHSP90 inhibitorCalcineurin inhibitor

Guanylate cyclase activator

LAQ82417-AAGCyclosporine

YC-1

Heterodimerization Antimicrobial agent Acriflavine
DNA binding AnthracyclineQuinoxaline antibiotic DoxorubicinEchinomycin
Transactivation Proteasome inhibitorAntifungal agent BortezomibAmphotericin B
Signal transduction BCR-ABL inhibitorCyclooxygenase inhibitorEGFR inhibitor

HER2 inhibitor

ImatinibIbuprofenErlotinib, Gefitinib

Trastuzumab

Over 100 women die every day of breast cancer in the U.S. The mean PO2 is 10 mm Hg in breast cancer as compared to > 60 mm Hg in normal breast tissue and cancers with PO2 < 10 mm Hg are associated with increased risk of metastasis and patient mortality (Vaupel et al., 2004). Increased HIF-1α protein levels, as identified by immunohistochemical analysis of tumor biopsies, are associated with increased risk of metastasis and/or patient mortality in unselected breast cancer patients and in lymph node-positive, lymph node-negative, HER2+, or estrogen receptor+ subpopulations (Semenza, 2011). Metastasis is responsible for > 90% of breast cancer mortality. The requirement for HIF-1 in breast cancer metastasis has been demonstrated for both autochthonous tumors in transgenic mice (Liao et al., 2007) and orthotopic transplants in immunodeficient mice (Zhang et al., 2011Wong et al., 2011). Primary tumors direct the recruitment of bone marrow-derived cells to the lungs and other sites of metastasis (Kaplan et al., 2005). In breast cancer, hypoxia induces the expression of lysyl oxidase (LOX), a secreted protein that remodels collagen at sites of metastatic niche formation (Erler et al., 2009). In addition to LOX, breast cancers also express LOX-like proteins 2 and 4. LOX, LOXL2, and LOXL4 are all HIF-1-regulated genes and HIF-1 inhibition blocks metastatic niche formation regardless of which LOX/LOXL protein is expressed, whereas available LOX inhibitors are not effective against all LOXL proteins (Wong et al., 2011), again illustrating the role of HIF-1 as a master regulator that controls the expression of multiple genes involved in a single (patho)physiological process.

Translational Prospects

Small molecule inhibitors of HIF activity that have anti-cancer effects in mouse models have been identified (Table 1). Inhibition of HIF impairs both vascular and metabolic adaptations to hypoxia, which may decrease O2 delivery and increase O2 utilization. These drugs are likely to be useful (as components of multidrug regimens) in the treatment of a subset of cancer patients in whom high HIF activity is driving progression. As with all novel cancer therapeutics, successful translation will require the development of methods for identifying the appropriate patient cohort. Effects of combination drug therapy also need to be considered. VEGF receptor tyrosine kinase inhibitors, which induce tumor hypoxia by blocking vascularization, have been reported to increase metastasis in mouse models (Ebos et al., 2009), which may be mediated by HIF-1; if so, combined use of HIF-1 inhibitors with these drugs may prevent unintended counter-therapeutic effects.

HIF inhibitors may also be useful in the treatment of other diseases in which dysregulated HIF activity is pathogenic. Proof of principle has been established in mouse models of ocular neovascularization, a major cause of blindness in the developed world, in which systemic or intraocular injection of the HIF-1 inhibitor digoxin is therapeutic (Yoshida et al., 2010). Systemic administration of HIF inhibitors for cancer therapy would be contraindicated in patients who also have ischemic cardiovascular disease, in which HIF activity is protective. The analysis of SNPs at the HIF1A locus described above suggests that the population may include HIF hypo-responders, who are at increased risk of severe ischemic cardiovascular disease. It is also possible that HIF hyper-responders, such as individuals with hereditary erythrocytosis, are at increased risk of particularly aggressive cancer.

O2 and Evolution, Part 2

When lowlanders sojourn to high altitude, hypobaric hypoxia induces erythropoiesis, which is a relatively ineffective response because the problem is not insufficient red cells, but rather insufficient ambient O2. Chronic erythrocytosis increases the risk of heart attack, stroke, and fetal loss during pregnancy. Many high-altitude Tibetans maintain the same hemoglobin concentration as lowlanders and yet, despite severe hypoxemia, they also maintain aerobic metabolism. The basis for this remarkable evolutionary adaptation appears to have involved the selection of genetic variants at multiple loci encoding components of the oxygen sensing system, particularly HIF-2α (Beall et al., 2010Simonson et al., 2010Yi et al., 2010). Given that hereditary erythrocytosis is associated with modest HIF-2α gain-of-function, the Tibetan genotype associated with absence of an erythrocytotic response to hypoxia may encode reduced HIF-2α activity along with other alterations that increase metabolic efficiency. Delineating the molecular mechanisms underlying these metabolic adaptations may lead to novel therapies for ischemic disorders, illustrating the importance of oxygen homeostasis as a nexus where evolution, biology, and medicine converge.

7.9.4 Hypoxia-inducible factor 1. Regulator of mitochondrial metabolism and mediator of ischemic preconditioning

Semenza GL1.
Biochim Biophys Acta. 2011 Jul; 1813(7):1263-8.
http://dx.doi.org/10.1016%2Fj.bbamcr.2010.08.006

Hypoxia-inducible factor 1 (HIF-1) mediates adaptive responses to reduced oxygen availability by regulating gene expression. A critical cell-autonomous adaptive response to chronic hypoxia controlled by HIF-1 is reduced mitochondrial mass and/or metabolism. Exposure of HIF-1-deficient fibroblasts to chronic hypoxia results in cell death due to excessive levels of reactive oxygen species (ROS). HIF-1 reduces ROS production under hypoxic conditions by multiple mechanisms including: a subunit switch in cytochrome c oxidase from the COX4-1 to COX4-2 regulatory subunit that increases the efficiency of complex IV; induction of pyruvate dehydrogenase kinase 1, which shunts pyruvate away from the mitochondria; induction of BNIP3, which triggers mitochondrial selective autophagy; and induction of microRNA-210, which blocks assembly of Fe/S clusters that are required for oxidative phosphorylation. HIF-1 is also required for ischemic preconditioning and this effect may be due in part to its induction of CD73, the enzyme that produces adenosine. HIF-1-dependent regulation of mitochondrial metabolism may also contribute to the protective effects of ischemic preconditioning.

The story of life on Earth is a tale of oxygen production and utilization. Approximately 3 billion years ago, primitive single-celled organisms evolved the capacity for photosynthesis, a biochemical process in which photons of solar energy are captured by chlorophyll and used to power the reaction of CO2 and H2O to form glucose and O2. The subsequent rise in the atmospheric O2 concentration over the next billion years set the stage for the ascendance of organisms with the capacity for respiration, a process that consumes glucose and O2 and generates CO2, H2O, and energy in the form of ATP. Some of these single-celled organisms eventually took up residence within the cytoplasm of other cells and devoted all of their effort to energy production as mitochondria. Compared to the conversion of glucose to lactate by glycolysis, the complete oxidation of glucose by respiration provided such a large increase in energy production that it made possible the evolution of multicellular organisms. Among metazoan organisms, the progressive increase in body size during evolution was accompanied by progressively more complex anatomic structures that function to ensure the adequate delivery of O2 to all cells, ultimately resulting in the sophisticated circulatory and respiratory systems of vertebrates.

All metazoan cells can sense and respond to reduced O2 availability (hypoxia). Adaptive responses to hypoxia can be cell autonomous, such as the alterations in mitochondrial metabolism that are described below, or non-cell-autonomous, such as changes in tissue vascularization (reviewed in ref. 1). Primary responses to hypoxia need to be distinguished from secondary responses to sequelae of hypoxia, such as the adaptive responses to ATP depletion that are mediated by AMP kinase (reviewed in ref 2). In contrast, recent data suggest that O2 and redox homeostasis are inextricably linked and that changes in oxygenation are inevitably associated with changes in the levels of reactive oxygen species (ROS), as will be discussed below.

HIF-1 Regulates Oxygen Homeostasis in All Metazoan Species

A key regulator of the developmental and physiological networks required for the maintenance of O2homeostasis is hypoxia-inducible factor 1 (HIF-1). HIF-1 is a heterodimeric transcription factor that is composed of an O2-regulated HIF-1α subunit and a constitutively expressed HIF-1β subunit [3,4]. HIF-1 regulates the expression of hundreds of genes through several major mechanisms. First, HIF-1 binds directly to hypoxia response elements, which are cis-acting DNA sequences located within target genes [5]. The binding of HIF-1 results in the recruitment of co-activator proteins that activate gene transcription (Fig. 1A). Only rarely does HIF-1 binding result in transcriptional repression [6]. Instead, HIF-1 represses gene expression by indirect mechanisms, which are described below. Second, among the genes activated by HIF-1 are many that encode transcription factors [7], which when synthesized can bind to and regulate (either positively or negatively) secondary batteries of target genes (Fig. 1B). Third, another group of HIF-1 target genes encode members of the Jumonji domain family of histone demethylases [8,9], which regulate gene expression by modifying chromatin structure (Fig. 1C). Fourth, HIF-1 can activate the transcription of genes encoding microRNAs [10], which bind to specific mRNA molecules and either block their translation or mediate their degradation (Fig. 1D). Fifth, the isolated HIF-1α subunit can bind to other transcription factors [11,12] and inhibit (Fig. 1E) or potentiate (Fig. 1F) their activity.

Mechanisms by which HIF-1 regulates gene expression. nihms232046f1

Mechanisms by which HIF-1 regulates gene expression. nihms232046f1

Mechanisms by which HIF-1 regulates gene expression.

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3010308/bin/nihms232046f1.gif

Fig. 1 Mechanisms by which HIF-1 regulates gene expression. (A) Top: HIF-1 binds directly to target genes at a cis-acting hypoxia response element (HRE) and recruits coactivator proteins such as p300 to increase gene transcription.

HIF-1α and HIF-1β are present in all metazoan species, including the simple roundworm Caenorhabitis elegans [13], which consists of ~103 cells and has no specialized systems for O2 delivery. The fruit flyDrosophila melanogaster evolved tracheal tubes, which conduct air into the interior of the body from which it diffuses to surrounding cells. In vertebrates, the development of the circulatory and respiratory systems was accompanied by the appearance of HIF-2α, which is also O2-regulated and heterodimerizes with HIF-1β [14] but is only expressed in a restricted number of cell types [15], whereas HIF-1α and HIF-1β are expressed in all human and mouse tissues [16]. In Drosophila, the ubiquitiously expressed HIF-1α ortholog is designatedSimilar [17] and the paralogous gene that is expressed specifically in tracheal tubes is designated Trachealess[18].

HIF-1 Activity is Regulated by Oxygen

In the presence of O2, HIF-1α and HIF-2α are subjected to hydroxylation by prolyl-4-hydroxylase domain proteins (PHDs) that use O2 and α-ketoglutarate as substrates and generate CO2 and succinate as by-products [19]. Prolyl hydroxylation is required for binding of the von Hipple-Lindau protein, which recruits a ubiquitin-protein ligase that targets HIF-1α and HIF-2α for proteasomal degradation (Fig. 2). Under hypoxic conditions, the rate of hydroxylation declines and the non-hydroxylated proteins accumulate. HIF-1α transactivation domain function is also O2-regulated [20,21]. Factor inhibiting HIF-1 (FIH-1) represses transactivation domain function [22] by hydroxylating asparagine residue 803 in HIF-1α, thereby blocking the binding of the co-activators p300 and CBP [23].

Negative regulation of HIF-1 activity by oxygen nihms232046f2

Negative regulation of HIF-1 activity by oxygen nihms232046f2

Negative regulation of HIF-1 activity by oxygen

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3010308/bin/nihms232046f2.gif

Fig. 2 Negative regulation of HIF-1 activity by oxygen. Top: In the presence of O2: prolyl hydroxylation of HIF-1a leads to binding of the von Hippel-Lindau protein (VHL), which recruits a ubiquitin protein-ligase that targets HIF-1a for proteasomal degradation;

When cells are acutely exposed to hypoxic conditions, the generation of ROS at complex III of the mitochondrial electron transport chain (ETC) increases and is required for the induction of HIF-1α protein levels [24]. More than a decade after these observations were first made, the precise mechanism by which hypoxia increases ROS generation and by which ROS induces HIF-1α accumulation remain unknown. However, the prolyl and asparaginyl hydroxylases contain Fe2+ in their active site and oxidation to Fe3+would block their catalytic activity. Since O2 is a substrate for the hydroxylation reaction, anoxia also results in a loss of enzyme activity. However, the concentration at which O2 becomes limiting for prolyl or asparaginyl hydroxylase activity in vivo is not known.

HIF-1 Regulates the Balance Between Oxidative and Glycolytic Metabolism

All metazoan organisms depend on mitochondrial respiration as the primary mechanism for generating sufficient amounts of ATP to maintain cellular and systemic homeostasis. Respiration, in turn, is dependent on an adequate supply of O2 to serve as the final electron acceptor in the ETC. In this process, electrons are transferred from complex I (or complex II) to complex III, then to complex IV, and finally to O2, which is reduced to water. This orderly transfer of electrons generates a proton gradient across the inner mitochondrial membrane that is used to drive the synthesis of ATP. At each step of this process, some electrons combine with O2 prematurely, resulting in the production of superoxide anion, which is reduced to hydrogen peroxide through the activity of mitochondrial superoxide dismutase. The efficiency of electron transport appears to be optimized to the physiological range of O2 concentrations, such that ATP is produced without the production of excess superoxide, hydrogen peroxide, and other ROS at levels that would result in the increased oxidation of cellular macromolecules and subsequent cellular dysfunction or death. In contrast, when O2levels are acutely increased or decreased, an imbalance between O2 and electron flow occurs, which results in increased ROS production.

MEFs require HIF-1 activity to make two critical metabolic adaptations to chronic hypoxia. First, HIF-1 activates the gene encoding pyruvate dehydrogenase (PDH) kinase 1 (PDK1), which phosphorylates and inactivates the catalytic subunit of PDH, the enzyme that converts pyruvate to acetyl coenzyme A (AcCoA) for entry into the mitochondrial tricarboxylic acid (TCA) cycle [25]. Second, HIF-1 activates the gene encoding BNIP3, a member of the Bcl-2 family of mitochondrial proteins, which triggers selective mitochondrial autophagy [26]. Interference with the induction of either of these proteins in hypoxic cells results in increased ROS production and increased cell death. Overexpression of either PDK1 or BNIP3 rescues HIF-1α-null MEFs. By shunting pyruvate away from the mitochondria, PDK1 decreases flux through the ETC and thereby counteracts the reduced efficiency of electron transport under hypoxic conditions, which would otherwise increase ROS production. PDK1 functions cooperatively with the product of another HIF-1 target gene, LDHA [27], which converts pyruvate to lactate, thereby further reducing available substrate for the PDH reaction.

PDK1 effectively reduces flux through the TCA cycle and thereby reduces flux through the ETC in cells that primarily utilize glucose as a substrate for oxidative phosphorylation. However, PDK1 is predicted to have little effect on ROS generation in cells that utilize fatty acid oxidation as their source of AcCoA. Hence another strategy to reduce ROS generation under hypoxic conditions is selective mitochondrial autophagy [26]. MEFs reduce their mitochondrial mass and O2 consumption by >50% after only two days at 1% O2. BNIP3 competes with Beclin-1 for binding to Bcl-2, thereby freeing Beclin-1 to activate autophagy. Using short hairpin RNAs to knockdown expression of BNIP3, Beclin-1, or Atg5 (another component of the autophagy machinery) phenocopied HIF-1α-null cells by preventing hypoxia-induced reductions in mitochondrial mass and O2 consumption as a result of failure to induce autophagy [26]. HIF-1-regulated expression of BNIP3L also contributes to hypoxia-induced autophagy [28]. Remarkably, mice heterozygous for the HIF-1α KO allele have a significantly increased ratio of mitochondrial:nuclear DNA in their lungs (even though this is the organ that is exposed to the highest O2 concentrations), indicating that HIF-1 regulates mitochondrial mass under physiological conditions in vivo [26]. In contrast to the selective mitochondrial autophagy that is induced in response to hypoxia as described above, autophagy (of unspecified cellular components) induced by anoxia does not require HIF-1, BNIP3, or BNIP3L, but is instead regulated by AMP kinase [29].

The multiplicity of HIF-1-mediated mechanisms identified so far by which cells regulate mitochondrial metabolism in response to changes in cellular O2 concentration (Fig. 3) suggests that this is a critical adaptive response to hypoxia. The fundamental nature of this physiological response is underscored by the fact that yeast also switch COX4 subunits in an O2-dependent manner but do so by an entirely different molecular mechanism [33], since yeast do not have a HIF-1α homologue. Thus, it appears that by convergent evolution both unicellular and multicellular eukaryotes possess mechanisms by which they modulate mitochondrial metabolism to maintain redox homeostasis despite changes in O2 availability. Indeed, it is the balance between energy, oxygen, and redox homeostasis that represents the key to life with oxygen.

Regulation of mitochondrial metabolism by HIF-1  nihms232046f3

Regulation of mitochondrial metabolism by HIF-1 nihms232046f3

Regulation of mitochondrial metabolism by HIF-1α

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3010308/bin/nihms232046f3.gif

Fig. 3 Regulation of mitochondrial metabolism by HIF-1α. Acute hypoxia leads to increased mitochondrial generation of reactive oxygen species (ROS). Decreased O2 and increased ROS levels lead to decreased HIF-1α hydroxylation (see Fig. 2) and increased HIF-1-dependent 

 

7.9.5 Regulation of cancer cell metabolism by hypoxia-inducible factor 1

Semenza GL1.
Semin Cancer Biol. 2009 Feb; 19(1):12-6.

The Warburg Effect: The Re-discovery of the Importance of Aerobic Glycolysis in Tumor Cells
http://dx.doi.org:/10.1016/j.semcancer.2008.11.009

The induction of hypoxia-inducible factor 1 (HIF-1) activity, either as a result of intratumoral hypoxia or loss-of-function mutations in the VHL gene, leads to a dramatic reprogramming of cancer cell metabolism involving increased glucose transport into the cell, increased conversion of glucose to pyruvate, and a concomitant decrease in mitochondrial metabolism and mitochondrial mass. Blocking these adaptive metabolic responses to hypoxia leads to cell death due to toxic levels of reactive oxygen species. Targeting HIF-1 or metabolic enzymes encoded by HIF-1 target genes may represent a novel therapeutic approach to cancer.

http://ars.els-cdn.com/content/image/1-s2.0-S1044579X08001065-gr1.sml

http://ars.els-cdn.com/content/image/1-s2.0-S1044579X08001065-gr2.sml

7.9.6 Coming up for air. HIF-1 and mitochondrial oxygen consumption

Simon MC1.
Cell Metab. 2006 Mar;3(3):150-1.
http://dx.doi.org/10.1016/j.cmet.2006.02.007

Hypoxic cells induce glycolytic enzymes; this HIF-1-mediated metabolic adaptation increases glucose flux to pyruvate and produces glycolytic ATP. Two papers in this issue of Cell Metabolism (Kim et al., 2006; Papandreou et al., 2006) demonstrate that HIF-1 also influences mitochondrial function, suppressing both the TCA cycle and respiration by inducing pyruvate dehydrogenase kinase 1 (PDK1). PDK1 regulation in hypoxic cells promotes cell survival.

Comment on

Oxygen deprivation (hypoxia) occurs in tissues when O2 supply via the cardiovascular system fails to meet the demand of O2-consuming cells. Hypoxia occurs naturally in physiological settings (e.g., embryonic development and exercising muscle), as well as in pathophysiological conditions (e.g., myocardial infarction, inflammation, and solid tumor formation). For over a century, it has been appreciated that O2-deprived cells exhibit increased conversion of glucose to lactate (the “Pasteur effect”). Activation of the Pasteur effect during hypoxia in mammalian cells is facilitated by HIF-1, which mediates the upregulation of glycolytic enzymes that support an increase in glycolytic ATP production as mitochondria become starved for O2, the substrate for oxidative phosphorylation (Seagroves et al., 2001). Thus, mitochondrial respiration passively decreases due to O2 depletion in hypoxic tissues. However, reports by Kim et al. (2006) and Papandreou et al. (2006) in this issue of Cell Metabolism demonstrate that this critical metabolic adaptation is more complex and includes an active suppression of mitochondrial pyruvate catabolism and O2consumption by HIF-1.

Mitochondrial oxidative phosphorylation is regulated by multiple mechanisms, including substrate availability. Major substrates include O2 (the terminal electron acceptor) and pyruvate (the primary carbon source). Pyruvate, as the end product of glycolysis, is converted to acetyl-CoA by the pyruvate dehydrogenase enzymatic complex and enters the tricarboxylic acid (TCA) cycle. Pyruvate conversion into acetyl-CoA is irreversible; this therefore represents an important regulatory point in cellular energy metabolism. Pyruvate dehydrogenase kinase (PDK) inhibits pyruvate dehydrogenase activity by phosphorylating its E1 subunit (Sugden and Holness, 2003). In the manuscripts by Kim et al. (2006) and Papandreou et al. (2006), the authors find that PDK1 is a HIF-1 target gene that actively regulates mitochondrial respiration by limiting pyruvate entry into the TCA cycle. By excluding pyruvate from mitochondrial metabolism, hypoxic cells accumulate pyruvate, which is then converted into lactate via lactate dehydrogenase (LDH), another HIF-1-regulated enzyme. Lactate in turn is released into the extracellular space, regenerating NAD+ for continued glycolysis by O2-starved cells (see Figure 1). This HIF-1-dependent block to mitochondrial O2 consumption promotes cell survival, especially when O2 deprivation is severe and prolonged.

multiple-hypoxia-induced-cellular-metabolic-changes-are-regulated-by-hif-1

multiple-hypoxia-induced-cellular-metabolic-changes-are-regulated-by-hif-1

http://ars.els-cdn.com/content/image/1-s2.0-S1550413106000672-gr1.jpg

Figure 1. Multiple hypoxia-induced cellular metabolic changes are regulated by HIF-1

By stimulating the expression of glucose transporters and glycolytic enzymes, HIF-1 promotes glycolysis to generate increased levels of pyruvate. In addition, HIF-1 promotes pyruvate reduction to lactate by activating lactate dehydrogenase (LDH). Pyruvate reduction to lactate regenerates NAD+, which permits continued glycolysis and ATP production by hypoxic cells. Furthermore, HIF-1 induces pyruvate dehydrogenase kinase 1 (PDK1), which inhibits pyruvate dehydrogenase and blocks conversion of pyruvate to acetyl CoA, resulting in decreased flux through the tricarboxylic acid (TCA) cycle. Decreased TCA cycle activity results in attenuation of oxidative phosphorylation and excessive mitochondrial reactive oxygen species (ROS) production. Because hypoxic cells already exhibit increased ROS, which have been shown to promote HIF-1 accumulation, the induction of PDK1 prevents the persistence of potentially harmful ROS levels.

Papandreou et al. demonstrate that hypoxic regulation of PDK has important implications for antitumor therapies. Recent interest has focused on cytotoxins that target hypoxic cells in tumor microenvironments, such as the drug tirapazamine (TPZ). Because intracellular O2 concentrations are decreased by mitochondrial O2 consumption, HIF-1 could protect tumor cells from TPZ-mediated cell death by maintaining intracellular O2 levels. Indeed, Papandreou et al. show that HIF-1-deficient cells grown at 2% O2 exhibit increased sensitivity to TPZ relative to wild-type cells, presumably due to higher rates of mitochondrial O2 consumption. HIF-1 inhibition in hypoxic tumor cells should have multiple therapeutic benefits, but the use of HIF-1 inhibitors in conjunction with other treatments has to be carefully evaluated for the most effective combination and sequence of drug delivery. One result of HIF-1 inhibition would be a relative decrease in intracellular O2 levels, making hypoxic cytotoxins such as TPZ more potent antitumor agents. Because PDK expression has been detected in multiple human tumor samples and appears to be induced by hypoxia (Koukourakis et al., 2005), small molecule inhibitors of HIF-1 combined with TPZ represent an attractive therapeutic approach for future clinical studies.

Hypoxic regulation of PDK1 has other important implications for cell survival during O2 depletion. Because the TCA cycle is coupled to electron transport, Kim et al. suggest that induction of the pyruvate dehydrogenase complex by PDK1 attenuates not only mitochondrial respiration but also the production of mitochondrial reactive oxygen species (ROS) in hypoxic cells. ROS are a byproduct of electron transfer to O2, and cells cultured at 1 to 5% O2 generate increased mitochondrial ROS relative to those cultured at 21% O2 (Chandel et al., 1998 and Guzy et al., 2005). In fact, hypoxia-induced mitochondrial ROS have also been shown to be necessary for the stabilization of HIF-1 in hypoxic cells (Brunelle et al., 2005Guzy et al., 2005 and Mansfield et al., 2005). However, the persistence of ROS could ultimately be lethal to tissues during chronic O2 deprivation, and PDK1 induction by HIF-1 should promote cell viability during long-term hypoxia. Kim et al. present evidence that HIF-1-deficient cells exhibit increased apoptosis after 72 hr of culture at 0.5% O2 compared to wild-type cells and that cell survival is rescued by enforced expression of exogenous PDK1. Furthermore, PDK1 reduces ROS production by the HIF-1 null cells. These findings support a novel prosurvival dimension of cellular hypoxic adaptation where PDK1 inhibits the TCA cycle, mitochondrial respiration, and chronic ROS production.

The HIF-1-mediated block to mitochondrial O2 consumption via PDK1 regulation also has implications for O2-sensing pathways by hypoxic cells. One school of thought suggests that perturbing mitochondrial O2consumption increases intracellular O2 concentrations and suppresses HIF-1 induction by promoting the activity of HIF prolyl hydroxylases, the O2-dependent enzymes that regulate HIF-1 stability (Hagen et al., 2003 and Doege et al., 2005). This model suggests that mitochondria function as “O2 sinks.” Although Papandreou et al. demonstrate that increased mitochondrial respiration due to PDK1 depletion results in decreased intracellular O2 levels (based on pimonidazole staining), these changes failed to reduce HIF-1 levels in hypoxic cells. Another model for hypoxic activation of HIF-1 describes a critical role for mitochondrial ROS in prolyl hydroxylase inhibition and HIF-1 stabilization in O2-starved cells (Brunelle et al., 2005Guzy et al., 2005 and Mansfield et al., 2005) (see Figure 1). The mitochondrial “O2 sink” hypothesis can account for some observations in the literature but fails to explain the inhibition of HIF-1 stabilization by ROS scavengers (Chandel et al., 1998Brunelle et al., 2005Guzy et al., 2005 and Sanjuán-Pla et al., 2005). While the relationship between HIF-1 stability, mitochondrial metabolism, ROS, and intracellular O2 redistribution will continue to be debated for some time, these most recent findings shed new light on findings by Louis Pasteur over a century ago.

Selected reading

Brunelle et al., 2005

J.K. Brunelle, E.L. Bell, N.M. Quesada, K. Vercauteren, V. Tiranti, M. Zeviani, R.C. Scarpulla, N.S. Chandel

Cell Metab., 1 (2005), pp. 409–414

Article  PDF (324 K) View Record in Scopus Citing articles (357)

Chandel et al., 1998

N.S. Chandel, E. Maltepe, E. Goldwasser, C.E. Mathieu, M.C. Simon, P.T. Schumacker

Proc. Natl. Acad. Sci. USA, 95 (1998), pp. 11715–11720

View Record in Scopus Full Text via CrossRef Citing articles (973)

Doege et al., 2005Doege, S. Heine, I. Jensen, W. Jelkmann, E. Metzen

Blood, 106 (2005), pp. 2311–2317

View Record in Scopus Full Text via CrossRef Citing articles (84)

Guzy et al., 2005

R.D. Guzy, B. Hoyos, E. Robin, H. Chen, L. Liu, K.D. Mansfield, M.C. Simon, U. Hammerling, P.T. Schumacker

Cell Metab., 1 (2005), pp. 401–408

Article  PDF (510 K) View Record in Scopus Citing articles (593)

Hagen et al., 2003

Hagen, C.T. Taylor, F. Lam, S. Moncada

Science, 302 (2003), pp. 1975–1978

View Record in Scopus Full Text via CrossRef Citing articles (450)

7.9.7 HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption

Papandreou I1Cairns RAFontana LLim ALDenko NC.
Cell Metab. 2006 Mar; 3(3):187-97.
http://dx.doi.org/10.1016/j.cmet.2006.01.012

The HIF-1 transcription factor drives hypoxic gene expression changes that are thought to be adaptive for cells exposed to a reduced-oxygen environment. For example, HIF-1 induces the expression of glycolytic genes. It is presumed that increased glycolysis is necessary to produce energy when low oxygen will not support oxidative phosphorylation at the mitochondria. However, we find that while HIF-1 stimulates glycolysis, it also actively represses mitochondrial function and oxygen consumption by inducing pyruvate dehydrogenase kinase 1 (PDK1). PDK1 phosphorylates and inhibits pyruvate dehydrogenase from using pyruvate to fuel the mitochondrial TCA cycle. This causes a drop in mitochondrial oxygen consumption and results in a relative increase in intracellular oxygen tension. We show by genetic means that HIF-1-dependent block to oxygen utilization results in increased oxygen availability, decreased cell death when total oxygen is limiting, and reduced cell death in response to the hypoxic cytotoxin tirapazamine.

Comment in

Tissue hypoxia results when supply of oxygen from the bloodstream does not meet demand from the cells in the tissue. Such a supply-demand mismatch can occur in physiologic conditions such as the exercising muscle, in the pathologic condition such as the ischemic heart, or in the tumor microenvironment (Hockel and Vaupel, 2001 and Semenza, 2004). In either the physiologic circumstance or pathologic conditions, there is a molecular response from the cell in which a program of gene expression changes is initiated by the hypoxia-inducible factor-1 (HIF-1) transcription factor. This program of gene expression changes is thought to help the cells adapt to the stressful environment. For example, HIF-1-dependent expression of erythropoietin and angiogenic compounds results in increased blood vessel formation for delivery of a richer supply of oxygenated blood to the hypoxic tissue. Additionally, HIF-1 induction of glycolytic enzymes allows for production of energy when the mitochondria are starved of oxygen as a substrate for oxidative phosphorylation. We now find that this metabolic adaptation is more complex, with HIF-1 not only regulating the supply of oxygen from the bloodstream, but also actively regulating the oxygen demand of the tissue by reducing the activity of the major cellular consumer of oxygen, the mitochondria.

Perhaps the best-studied example of chronic hypoxia is the hypoxia associated with the tumor microenvironment (Brown and Giaccia, 1998). The tumor suffers from poor oxygen supply through a chaotic jumble of blood vessels that are unable to adequately perfuse the tumor cells. The oxygen tension within the tumor is also a function of the demand within the tissue, with oxygen consumption influencing the extent of tumor hypoxia (Gulledge and Dewhirst, 1996 and Papandreou et al., 2005b). The net result is that a large fraction of the tumor cells are hypoxic. Oxygen tensions within the tumor range from near normal at the capillary wall, to near zero in the perinecrotic regions. This perfusion-limited hypoxia is a potent microenvironmental stress during tumor evolution (Graeber et al., 1996 and Hockel and Vaupel, 2001) and an important variable capable of predicting for poor patient outcome. (Brizel et al., 1996Cairns and Hill, 2004Hockel et al., 1996 and Nordsmark and Overgaard, 2004).

The HIF-1 transcription factor was first identified based on its ability to activate the erythropoetin gene in response to hypoxia (Wang and Semenza, 1993). Since then, it is has been shown to be activated by hypoxia in many cells and tissues, where it can induce hypoxia-responsive target genes such as VEGF and Glut1 (Airley et al., 2001 and Kimura et al., 2004). The connection between HIF-regulation and human cancer was directly linked when it was discovered that the VHL tumor suppressor gene was part of the molecular complex responsible for the oxic degradation of HIF-1α (Maxwell et al., 1999). In normoxia, a family of prolyl hydroxylase enzymes uses molecular oxygen as a substrate and modifies HIF-1α and HIF2α by hydroxylation of prolines 564 and 402 (Bruick and McKnight, 2001 and Epstein et al., 2001). VHL then recognizes the modified HIF-α proteins, acts as an E3-type of ubiquitin ligase, and along with elongins B and C is responsible for the polyubiquitination of HIF-αs and their proteosomal degradation (Bruick and McKnight, 2001Chan et al., 2002Ivan et al., 2001 and Jaakkola et al., 2001). Mutations in VHL lead to constitutive HIF-1 gene expression, and predispose humans to cancer. The ability to recognize modified HIF-αs is at least partly responsible for VHL activity as a tumor suppressor, as introduction of nondegradable HIF-2α is capable of overcoming the growth–inhibitory activity of wild-type (wt) VHL in renal cancer cells (Kondo et al., 2003).

Mitochondrial function can be regulated by PDK1 expression. Mitochondrial oxidative phosphorylation (OXPHOS) is regulated by several mechanisms, including substrate availability (Brown, 1992). The major substrates for OXPHOS are oxygen, which is the terminal electron acceptor, and pyruvate, which is the primary carbon source. Pyruvate is the end product of glycolysis and is converted to acetyl-CoA through the activity of the pyruvate dehydrogenase complex of enzymes. The acetyl-CoA then directly enters the TCA cycle at citrate synthase where it is combined with oxaloacetate to generate citrate. In metazoans, the conversion of pyruvate to acetyl-CoA is irreversible and therefore represents a critical regulatory point in cellular energy metabolism. Pyruvate dehydrogenase is regulated by three known mechanisms: it is inhibited by acetyl-CoA and NADH, it is stimulated by reduced energy in the cell, and it is inhibited by regulatory phosphorylation of its E1 subunit by pyruvate dehydrogenase kinase (PDK) (Holness and Sugden, 2003 and Sugden and Holness, 2003). There are four members of the PDK family in vertebrates, each with specific tissue distributions (Roche et al., 2001). PDK expression has been observed in human tumor biopsies (Koukourakis et al., 2005), and we have reported that PDK3 is hypoxia-inducible in some cell types (Denko et al., 2003). In this manuscript, we find that PDK1 is also a hypoxia-responsive protein that actively regulates the function of the mitochondria under hypoxic conditions by reducing pyruvate entry into the TCA cycle. By excluding pyruvate from mitochondrial consumption, PDK1 induction may increase the conversion of pyruvate to lactate, which is in turn shunted to the extracellular space, regenerating NAD for continued glycolysis.

Identification of HIF-dependent mitochondrial proteins through genomic and bioinformatics approaches

In order to help elucidate the role of HIF-1α in regulating metabolism, we undertook a genomic search for genes that were regulated by HIF-1 in tumor cells exposed to hypoxia in vitro. We used genetically matched human RCC4 cells that had lost VHL during tumorigenesis and displayed constitutive HIF-1 activity, and a cell line engineered to re-express VHL to establish hypoxia-dependent HIF activation. These cells were treated with 18 hr of stringent hypoxia (<0.01% oxygen), and microarray analysis performed. Using a strict 2.5-fold elevation as our cutoff, we identified 173 genes that were regulated by hypoxia and/or VHL status (Table S1 in the Supplemental Data available with this article online). We used the pattern of expression in these experiments to identify putative HIF-regulated genes—ones that were constitutively elevated in the parent RCC4s independent of hypoxia, downregulated in the RCC4VHL cells under normoxia, and elevated in response to hypoxia. Of the 173 hypoxia and VHL-regulated genes, 74 fit the putative HIF-1 target pattern. The open reading frames of these genes were run through a pair of bioinformatics engines in order to predict subcellular localization, and 10 proteins scored as mitochondrial on at least one engine. The genes, fold induction, and mitochondrial scores are listed in Table 1.

HIF-1 downregulates mitochondrial oxygen consumption

Having identified several putative HIF-1 responsive gene products that had the potential to regulate mitochondrial function, we then directly measured mitochondrial oxygen consumption in cells exposed to long-term hypoxia. While other groups have studied mitochondrial function under acute hypoxia (Chandel et al., 1997), this is one of the first descriptions of mitochondrial function after long-term hypoxia where there have been extensive hypoxia-induced gene expression changes. Figure 1A is an example of the primary oxygen trace from a Clark electrode showing a drop in oxygen concentration in cell suspensions of primary fibroblasts taken from normoxic and hypoxic cultures. The slope of the curve is a direct measure of the total cellular oxygen consumption rate. Exposure of either primary human or immortalized mouse fibroblasts to 24 hr of hypoxia resulted in a reduction of this rate by approximately 50% (Figures 1A and 1B). In these experiments, the oxygen consumption can be stimulated with the mitochondrial uncoupling agent CCCP (carbonyl cyanide 3-chloro phenylhydrazone) and was completely inhibited by 2 mM potassium cyanide. We determined that the change in total cellular oxygen consumption was due to changes in mitochondrial activity by the use of the cell-permeable poison of mitochondrial complex 3, Antimycin A. Figure 1C shows that the difference in the normoxic and hypoxic oxygen consumption in murine fibroblasts is entirely due to the Antimycin-sensitive mitochondrial consumption. The kinetics with which mitochondrial function slows in hypoxic tumor cells also suggests that it is due to gene expression changes because it takes over 6 hr to achieve maximal reduction, and the reversal of this repression requires at least another 6 hr of reoxygenation (Figure 1D). These effects are not likely due to proliferation or toxicity of the treatments as these conditions are not growth inhibitory or toxic to the cells (Papandreou et al., 2005a).

Since we had predicted from the gene expression data that the mitochondrial oxygen consumption changes were due to HIF-1-mediated expression changes, we tested several genetically matched systems to determine what role HIF-1 played in the process (Figure 2). We first tested the cell lines that had been used for microarray analysis and found that the parental RCC4 cells had reduced mitochondrial oxygen consumption when compared to the VHL-reintroduced cells. Oxygen consumption in the parental cells was insensitive to hypoxia, while it was reduced by hypoxia in the wild-type VHL-transfected cell lines. Interestingly, stable introduction of a tumor-derived mutant VHL (Y98H) that cannot degrade HIF was also unable to restore oxygen consumption. These results indicate that increased expression of HIF-1 is sufficient to reduce oxygen consumption (Figure 2A). We also investigated whether HIF-1 induction was required for the observed reduction in oxygen consumption in hypoxia using two genetically matched systems. We measured normoxic and hypoxic oxygen consumption in murine fibroblasts derived from wild-type or HIF-1α null embryos (Figure 2B) and from human RKO tumor cells and RKO cells constitutively expressing ShRNAs directed against the HIF-1α gene (Figures 2C and 4C). Neither of the HIF-deficient cell systems was able to reduce oxygen consumption in response to hypoxia. These data from the HIF-overexpressing RCC cells and the HIF-deficient cells indicate that HIF-1 is both necessary and sufficient for reducing mitochondrial oxygen consumption in hypoxia.

HIF-dependent mitochondrial changes are functional, not structural

Because addition of CCCP could increase oxygen consumption even in the hypoxia-treated cells, we hypothesized that the hypoxic inhibition was a regulated activity, not a structural change in the mitochondria in response to hypoxic stress. We confirmed this interpretation by examining several additional mitochondrial characteristics in hypoxic cells such as mitochondrial morphology, quantity, and membrane potential. We examined morphology by visual inspection of both the transiently transfected mitochondrially localized DsRed protein and the endogenous mitochondrial protein cytochrome C. Both markers were indistinguishable in the parental RCC4 and the RCC4VHL cells (Figure 3A). Likewise, we measured the mitochondrial membrane potential with the functional dye rhodamine 123 and found that it was identical in the matched RCC4 cells and the matched HIF wt and knockout (KO) cells when cultured in normoxia or hypoxia (Figure 3B). Finally, we determined that the quantity of mitochondria per cell was not altered in response to HIF or hypoxia by showing that the amount of the mitochondrial marker protein HSP60 was identical in the RCC4 and HIF cell lines (Figure 3C)

PDK1 is a HIF-1 inducible target protein

After examination of the list of putative HIF-regulated mitochondrial target genes, we hypothesized that PDK1 could mediate the functional changes that we observed in hypoxia. We therefore investigated PDK1 protein expression in response to HIF and hypoxia in the genetically matched cell systems. Figure 4A shows that in the RCC4 cells PDK1 and the HIF-target gene BNip3 (Greijer et al., 2005 and Papandreou et al., 2005a) were both induced by hypoxia in a VHL-dependent manner, with the expression of PDK1 inversely matching the oxygen consumption measured in Figure 1 above. Likewise, the HIF wt MEFs show oxygen-dependent induction of PDK1 and BNip3, while the HIF KO MEFs did not show any expression of either of these proteins under any oxygen conditions (Figure 4B). Finally, the parental RKO cells were able to induce PDK1 and the HIF target gene BNip3L in response to hypoxia, while the HIF-depleted ShRNA RKO cells could not induce either protein (Figure 4C). Therefore, in all three cell types, the HIF-1-dependent regulation of oxygen consumption seen in Figure 2, corresponds to the HIF-1-dependent induction of PDK1 seen in Figure 4.

In order to determine if PDK1 was a direct HIF-1 target gene, we analyzed the genomic sequence flanking the 5′ end of the gene for possible HIF-1 binding sites based on the consensus core HRE element (A/G)CGTG (Caro, 2001). Several such sites exist within the first 400 bases upstream, so we generated reporter constructs by fusing the genomic sequence from −400 to +30 of the start site of transcription to the firefly luciferase gene. In transfection experiments, the chimeric construct showed significant induction by either cotransfection with a constitutively active HIF proline mutant (P402A/P564G) (Chan et al., 2002) or exposure of the transfected cells to 0.5% oxygen (Figure 4D). Most noteworthy, when the reporter gene was transfected into the HIF-1α null cells, it did not show induction when the cells were cultured in hypoxia, but it did show induction when cotransfected with expression HIF-1α plasmid. We then generated deletions down to the first 36 bases upstream of transcription and found that even this short sequence was responsive to HIF-1 (Figure 4D). Analysis of this small fragment showed only one consensus HRE site located in an inverted orientation in the 5′ untranslated region. We synthesized and cloned a mutant promoter fragment in which the core element ACGTG was replaced with AAAAG, and this construct lost over 90% of its hypoxic induction. These experiments suggest that it is this HRE within the proximal 5′ UTR that HIF-1 uses to transactivate the endogenous PDK1 gene in response to hypoxia.

PDK1 is responsible for the HIF-dependent mitochondrial oxygen consumption changes

In order to directly test if PDK1 was the HIF-1 target gene responsible for the hypoxic reduction in mitochondrial oxygen consumption, we generated RKO cell lines with either knockdown or overexpression of PDK1 and measured the oxygen consumption in these derivatives. The PDK1 ShRNA stable knockdown line was generated as a pool of clones cotransfected with pSUPER ShPDK1 and pTK-hygro resistance gene. After selection for growth in hygromycin, the cells were tested by Western blot for the level of PDK1 protein expression. We found that normoxic PDK1 is reduced by 75%, however, there was measurable expression of PDK1 in these cells in response to hypoxia (Figure 5A). When we measured the corresponding oxygen consumption in these cells, we found a change commensurate with the level of PDK1. The knockdown cells show elevated baseline oxygen consumption, and partial reduction in this activity in response to hypoxia. Therefore, reduction of PDK1 expression by genetic means increased mitochondrial oxygen consumption in both normoxic and hypoxic conditions. Interestingly, these cells still induced HIF-1α (Figure 5A) and HIF-1 target genes such as BNip3L in response to hypoxia (data not shown), suggesting that altered PDK1 levels do not alter HIF-1α function.

pdk1-expression-directly-regulates-cellular-oxygen-consumption-rate

pdk1-expression-directly-regulates-cellular-oxygen-consumption-rate

PDK1 expression directly regulates cellular oxygen consumption rate

http://ars.els-cdn.com/content/image/1-s2.0-S155041310600060X-gr5.jpg

Figure 5. PDK1 expression directly regulates cellular oxygen consumption rate

  1. A)Western blot of RKO cell and ShRNAPDK1RKO cell lysates after exposure to 24 hr of normoxia or 0.5% O2. Blots were probed for HIF 1α, PDK1, and tubulin as a loading control.
  2. B)Oxygen consumption rate in RKO and ShRNAPDK1RKO cells after exposure to 24 hr of normoxia or 0.5% O2.
  3. C)Western blot of RKOiresGUS cell and RKOiresPDK1 cell lysates after exposure to 24 hr of normoxia or 0.5% O2. Blots were probed for HIF 1α, PDK1, and tubulin as a loading control.
  4. D)Oxygen consumption rate in RKOiresGUS and RKOiresPDK1 cells after exposure to 24 hr of normoxia or 0.5% O2.
  5. E)Model describing the interconnected effects of HIF-1 target gene activation on hypoxic cell metabolism. Reduced oxygen conditions causes HIF-1 to coordinately induce the enzymes shown in boxes. HIF-1 activation results in increased glucose transporter expression to increase intracellular glucose flux, induction of glycolytic enzymes increases the conversion of glucose to pyruvate generating energy and NADH, induction of PDK1 decreases mitochondrial utilization of pyruvate and oxygen, and induction of LDH increases the removal of excess pyruvate as lactate and also regenerates NAD+ for increased glycolysis.

For all graphs, the error bars represent the standard error of the mean.

We also determined if overexpression of PDK1 could lead to reduced mitochondrial oxygen consumption. A separate culture of RKO cells was transfected with a PDK1-IRES-puro expression plasmid and selected for resistance to puromycin. The pool of puromycin resistant cells was tested for PDK1 expression by Western blot. These cells showed a modest increase in PDK1 expression under control conditions when compared to the cells transfected with GUS-IRES-puro, with an additional increase in PDK1 protein in response to hypoxia (Figure 5C). The corresponding oxygen consumption measurements showed that the mitochondria is very sensitive to changes in the levels of PDK1, as even this slight increase was able to significantly reduce oxygen consumption in the normoxic PDK1-puro cultures. Further increase in PDK1 levels with hypoxia further reduced oxygen consumption in both cultures (Figure 5D). The model describing the relationship between hypoxia, HIF-1, PDK1, and intermediate metabolism is described inFigure 5E.

Altering oxygen consumption alters intracellular oxygen tension and sensitivity to hypoxia-dependent cell killing

The intracellular concentration of oxygen is a net result of the rate at which oxygen diffuses into the cell and the rate at which it is consumed. We hypothesized that the rate at which oxygen was consumed within the cell would significantly affect its steady-state intracellular concentrations. We tested this hypothesis in vitro using the hypoxic marker drug pimonidazole (Bennewith and Durand, 2004). We plated high density cultures of HIF wild-type and HIF knockout cells and placed these cultures in normoxic, 2% oxygen, and anoxic incubators for overnight treatment. The overnight treatment gives the cells time to adapt to the hypoxic conditions and establish altered oxygen consumption profiles. Pimonidozole was then added for the last 4 hr of the growth of the culture. Pimonidazole binding was detected after fixation of the cells using an FITC labeled anti-pimonidazole antibody and it was quantitated by flow cytometry. The quantity of the bound drug is a direct indication of the oxygen concentration within the cell (Bennewith and Durand, 2004). The histograms in Figure 6A show that the HIF-1 knockout and wild-type cells show similar staining in the cells grown in 0% oxygen. However, the cells treated with 2% oxygen show the consequence of the genetic removal of HIF-1. The HIF-proficient cells showed relatively less pimonidazole binding at 2% when compared to the 0% culture, while the HIF-deficient cells showed identical binding between the cells at 2% and those at 0%. We interpret these results to mean that the HIF-deficient cells have greater oxygen consumption, and this has lowered the intracellular oxygenation from the ambient 2% to close to zero intracellularly. The HIF-proficient cells reduced their oxygen consumption rate so that the rate of diffusion into the cell is greater than the rate of consumption.

Figure 6. HIF-dependent decrease in oxygen consumption raises intracellular oxygen concentration, protects when oxygen is limiting, and decreases sensitivity to tirapazamine in vitro

  1. A)Pimonidazole was used to determine the intracellular oxygen concentration of cells in culture. HIF wt and HIF KO MEFs were grown at high density and exposed to 2% O2or anoxia for 24 hr in glass dishes. For the last 4 hr of treatment, cells were exposed to 60 μg/ml pimonidazole. Pimonidazole binding was quantitated by flow cytometry after binding of an FITC conjugated anti-pimo mAb. Results are representative of two independent experiments.
  2. B)HIF1α reduces oxygen consumption and protects cells when total oxygen is limited. HIF wt and HIF KO cells were plated at high density and sealed in aluminum jigs at <0.02% oxygen. At the indicated times, cells were harvested, and dead cells were quantitated by trypan blue exclusion. Note both cell lines are equally sensitive to anoxia-induced apoptosis, so the death of the HIF null cells indicates that the increased oxygen consumption removed any residual oxygen in the jig and resulted in anoxia-induced death.
  3. C)PDK1 is responsible for HIF-1’s adaptive response when oxygen is limiting. A similar jig experiment was performed to measure survival in the parental RKO, the RKO ShRNAHIF1α, and the RKOShPDK1 cells. Cell death by trypan blue uptake was measured 48 hr after the jigs were sealed.
  4. D)HIF status alters sensitivity to TPZ in vitro. HIF wt and HIF KO MEFs were grown at high density in glass dishes and exposed to 21%, 2%, and <0.01% O2conditions for 18 hr in the presence of varying concentrations of Tirapazamine. After exposure, cells were harvested and replated under normoxia to determine clonogenic viability. Survival is calculated relative to the plating efficiency of cells exposed to 0 μM TPZ for each oxygen concentration.
  5. E)Cell density alters sensitivity to TPZ. HIF wt and HIF KO MEFs were grown at varying cell densities in glass dishes and exposed to 2% O2in the presence of 10 μM TPZ for 18 hr. After the exposure, survival was determined as described in (C).

For all graphs, the error bars represent the standard error of the mean.

HIF-induced PDK1 can reduce the total amount of oxygen consumed per cell. The reduction in the amount of oxygen consumed could be significant if there is a finite amount of oxygen available, as would be the case in the hours following a blood vessel occlusion. The tissue that is fed by the vessel would benefit from being economical with the oxygen that is present. We experimentally modeled such an event using aluminum jigs that could be sealed with defined amounts of cells and oxygen present (Siim et al., 1996). We placed 10 × 106 wild-type or HIF null cells in the sealed jig at 0.02% oxygen, waited for the cells to consume the remaining oxygen, and measured cell viability. We have previously shown that these two cell types are resistant to mild hypoxia and equally sensitive to anoxia-induced apoptosis (Papandreou et al., 2005a). Therefore, any death in this experiment would be the result of the cells consuming the small amount of remaining oxygen and dying in response to anoxia. We found that in sealed jigs, the wild-type cells are more able to adapt to the limited oxygen supply by reducing consumption. The HIF null cells continued to consume oxygen, reached anoxic levels, and started to lose viability within 36 hr (Figure 6B). This is a secondary adaptive effect of HIF1. We confirmed that PDK1 was responsible for this difference by performing a similar experiment using the parental RKO cells, the RKOShRNAHIF1α and the RKOShRNAPDK1 cells. We found similar results in which both the cells with HIF1α knockdown and PDK1 knockdown were sensitive to the long-term effects of being sealed in a jig with a defined amount of oxygen (Figure 6c). Note that the RKOShPDK1 cells are even more sensitive than the RKOShHIF1α cells, presumably because they have higher basal oxygen consumption rates (Figure 5B).

Because HIF-1 can help cells adapt to hypoxia and maintain some intracellular oxygen level, it may also protect tumor cells from killing by the hypoxic cytotoxin tirapazamine (TPZ). TPZ toxicity is very oxygen dependent, especially at oxygen levels between 1%–4% (Koch, 1993). We therefore tested the relative sensitivity of the HIF wt and HIF KO cells to TPZ killing in high density cultures (Figure 6D). We exposed the cells to the indicated concentrations of drug and oxygen concentrations overnight. The cells were then harvested and replated to determine reproductive viability by colony formation. Both cell types were equally resistant to TPZ at 21% oxygen, while both cell types are equally sensitive to TPZ in anoxic conditions where intracellular oxygen levels are equivalent (Figure 6A). The identical sensitivity of both cell types in anoxia indicates that both cell types are equally competent in repairing the TPZ-induced DNA damage that is presumed to be responsible for its toxicity. However, in 2% oxygen cultures, the HIF null cells displayed a significantly greater sensitivity to the drug than the wild-type cells. This suggests that the increased oxygen consumption rate in the HIF-deficient cells is sufficient to lower the intracellular oxygen concentration relative to that in the HIF-proficient cells. The lower oxygen level is significant enough to dramatically sensitize these cells to killing by TPZ.

If the increased sensitivity to TPZ in the HIF ko cells is determined by intracellular oxygen consumption differences, then this effect should also be cell-density dependent. We showed that this is indeed the case in Figure 6E where oxygen and TPZ concentrations were held constant, and increased cell density lead to increased TPZ toxicity. The effect was much more pronounced in the HIF KO cells, although the HIF wt cells showed some increased toxicity in the highest density cultures, consistent with the fact they were still consuming some oxygen, even with HIF present (Figure 1). The in vitro TPZ survival data is therefore consistent with our hypothesis that control of oxygen consumption can regulate intracellular oxygen concentration, and suggests that increased oxygen consumption could sensitize cells to hypoxia-dependent therapy.

Discussion

The findings presented here show that HIF-1 is actively responsible for regulating energy production in hypoxic cells by an additional, previously unrecognized mechanism. It has been shown that HIF-1 induces the enzymes responsible for glycolysis when it was presumed that low oxygen did not support efficient oxidative phosphorylation (Iyer et al., 1998 and Seagroves et al., 2001). The use of glucose to generate ATP is capable of satisfying the energy requirements of a cell if glucose is in excess (Papandreou et al., 2005a). We now find that at the same time that glycolysis is increasing, mitochondrial respiration is decreasing. However, the decreased respiration is not because there is not enough oxygen present to act as a substrate for oxidative phosphorylation, but because the flow of pyruvate into the TCA cycle has been reduced by the activity of pyruvate dehydrogenase kinase. Other reports have suggested that oxygen utilization is shifted in cells exposed to hypoxia, but these reports have focused on other regulators such as nitric oxide synthase (Hagen et al., 2003). NO can reduce oxygen consumption through direct inhibition of cytochrome oxidase, but this effect seems to be more significant at physiologic oxygen concentrations, not at severe levels seen in the tumor (Palacios-Callender et al., 2004).

7.9.8 HIF-1. upstream and downstream of cancer metabolism

Semenza GL1.
Curr Opin Genet Dev. 2010 Feb; 20(1):51-6
http://dx.doi.org/10.1016%2Fj.gde.2009.10.009

Hypoxia-inducible factor 1 (HIF-1) plays a key role in the reprogramming of cancer metabolism by activating transcription of genes encoding glucose transporters and glycolytic enzymes, which take up glucose and convert it to lactate; pyruvate dehydrogenase kinase 1, which shunts pyruvate away from the mitochondria; and BNIP3, which triggers selective mitochondrial autophagy. The shift from oxidative to glycolytic metabolism allows maintenance of redox homeostasis and cell survival under conditions of prolonged hypoxia. Many metabolic abnormalities in cancer cells increase HIF-1 activity. As a result, a feed-forward mechanism can be activated that drives HIF-1 activation and may promote tumor progression. Hypoxia-inducible factor 1 (HIF-1) plays a key role in the reprogramming of cancer metabolism by activating transcription of genes encoding glucose transporters and glycolytic enzymes, which take up glucose and convert it to lactate; pyruvate dehydrogenase kinase 1, which shunts pyruvate away from the mitochondria; and BNIP3, which triggers selective mitochondrial autophagy. The shift from oxidative to glycolytic metabolism allows maintenance of redox homeostasis and cell survival under conditions of prolonged hypoxia. Many metabolic abnormalities in cancer cells increase HIF-1 activity. As a result, a feed-forward mechanism can be activated that drives HIF-1 activation and may promote tumor progression.

Metastatic cancer is characterized by reprogramming of cellular metabolism leading to increased uptake of glucose for use as both an anabolic and catabolic substrate. Increased glucose uptake is such a reliable feature that it is utilized clinically to detect metastases by positron emission tomography using 18F-fluorodeoxyglucose (FDG-PET) with a sensitivity of ~90% [1]. As with all aspects of cancer biology, the details of metabolic reprogramming differ widely among individual tumors. However, the role of specific signaling pathways and transcription factors in this process is now understood in considerable detail. This review will focus on the involvement of hypoxia-inducible factor 1 (HIF-1) in both mediating metabolic reprogramming and responding to metabolic alterations. The placement of HIF-1 both upstream and downstream of cancer metabolism results in a feed-forward mechanism that may play a major role in the development of the invasive, metastatic, and lethal cancer phenotype.

O2 concentrations are significantly reduced in many human cancers compared to the surrounding normal tissue. The median PO2 in breast cancers is ~10 mm Hg, as compared to ~65 mm Hg in normal breast tissue [2]. Reduced O2 availability induces HIF-1, which regulates the transcription of hundreds of genes [3*,4*] that encode proteins involved in every aspect of cancer biology, including: cell immortalization and stem cell maintenance; genetic instability; glucose and energy metabolism; vascularization; autocrine growth factor signaling; invasion and metastasis; immune evasion; and resistance to chemotherapy and radiation therapy [5].

HIF-1 is a transcription factor that consists of an O2-regulated HIF-1α and a constitutively expressed HIF-1β subunit [6]. In well-oxygenated cells, HIF-1α is hydroxylated on proline residue 402 (Pro-402) and/or Pro-564 by prolyl hydroxylase domain protein 2 (PHD2), which uses O2 and α-ketoglutarate as substrates in a reaction that generates CO2 and succinate as byproducts [7]. Prolyl-hydroxylated HIF-1α is bound by the von Hippel-Lindau tumor suppressor protein (VHL), which recruits an E3-ubiquitin ligase that targets HIF-1α for proteasomal degradation (Figure 1A). Asparagine 803 in the transactivation domain is hydroxylated in well-oxygenated cells by factor inhibiting HIF-1 (FIH-1), which blocks the binding of the coactivators p300 and CBP [7]. Under hypoxic conditions, the prolyl and asparaginyl hydroxylation reactions are inhibited by substrate (O2) deprivation and/or the mitochondrial generation of reactive oxygen species (ROS), which may oxidize Fe(II) present in the catalytic center of the hydroxylases [8].

HIF-1 and metabolism  nihms156580f1

HIF-1 and metabolism nihms156580f1

HIF-1 and metabolism

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2822127/bin/nihms156580f1.gif

Figure 1 HIF-1 and metabolism. (A) Regulation of HIF-1α protein synthesis and stability and HIF-1-dependent metabolic reprogramming. The rate of translation of HIF-1α mRNA into protein in cancer cells is dependent upon the activity of the mammalian 

The finding that acute changes in PO2 increase mitochondrial ROS production suggests that cellular respiration is optimized at physiological PO2 to limit ROS generation and that any deviation in PO2 — up or down — results in increased ROS generation. If hypoxia persists, induction of HIF-1 leads to adaptive mechanisms to reduce ROS and re-establish homeostasis, as described below. Prolyl and asparaginyl hydroxylation provide a molecular mechanism by which changes in cellular oxygenation can be transduced to the nucleus as changes in HIF-1 activity. This review will focus on recent advances in our understanding of the role of HIF-1 in controlling glucose and energy metabolism, but it should be appreciated that any increase in HIF-1 activity that leads to changes in cell metabolism will also affect many other critical aspects of cancer biology [5] that will not be addressed here.

HIF-1 target genes involved in glucose and energy metabolism

HIF-1 activates the transcription of SLC2A1 and SLC2A3, which encode the glucose transporters GLUT1 and GLUT3, respectively, as well as HK1 and HK2, which encode hexokinase, the first enzyme of the Embden-Meyerhoff (glycolytic) pathway [9]. Once taken up by GLUT and phosphorylated by HK, FDG cannot be metabolized further; thus, FDG-PET signal is determined by FDG delivery to tissue (i.e. perfusion) and GLUT/HK expression/activity. Unlike FDG, glucose is further metabolized to pyruvate by the action of the glycolytic enzymes, which are all encoded by HIF-1 target genes (Figure 1A). Glycolytic intermediates are also utilized for nucleotide and lipid synthesis [10]. Lactate dehydrogenase A (LDHA), which converts pyruvate to lactate, and monocarboxylate transporter 4 (MCT4), which transports lactate out of the cell (Figure 1B), are also regulated by HIF-1 [9,11]. Remarkably, lactate produced by hypoxic cancer cells can be taken up by non-hypoxic cells and used as a respiratory substrate [12**].

Pyruvate represents a critical metabolic control point, as it can be converted to acetyl coenzyme A (AcCoA) by pyruvate dehydrogenase (PDH) for entry into the tricarboxylic acid (TCA) cycle or it can be converted to lactate by LDHA (Figure 1B). Pyruvate dehydrogenase kinase (PDK), which phosphorylates and inactivates the catalytic domain of PDH, is encoded by four genes and PDK1 is activated by HIF-1 [13,14]. (Further studies are required to determine whether PDK2PDK3, or PDK4 is regulated by HIF-1.) As a result of PDK1 activation, pyruvate is actively shunted away from the mitochondria, which reduces flux through the TCA cycle, thereby reducing delivery of NADH and FADH2 to the electron transport chain. This is a critical adaptive response to hypoxia, because in HIF-1α–null mouse embryo fibroblasts (MEFs), PDK1 expression is not induced by hypoxia and the cells die due to excess ROS production, which can be ameliorated by forced expression of PDK1 [13]. MYC, which is activated in ~40% of human cancers, cooperates with HIF-1 to activate transcription of PDK1, thereby amplifying the hypoxic response [15]. Pharmacological inhibition of HIF-1 or PDK1 activity increases O2 consumption by cancer cells and increases the efficacy of a hypoxia-specific cytotoxin [16].

Hypoxia also induces mitochondrial autophagy in many human cancer cell lines through HIF-1-dependent expression of BNIP3 and a related BH3 domain protein, BNIP3L [19**]. Autocrine signaling through the platelet-derived growth factor receptor in cancer cells increases HIF-1 activity and thereby increases autophagy and cell survival under hypoxic conditions [21]. Autophagy may also occur in a HIF-1-independent manner in response to other physiological stimuli that are associated with hypoxic conditions, such as a decrease in the cellular ATP:AMP ratio, which activates AMP kinase signaling [22].

In clear cell renal carcinoma, VHL loss of function (LoF) results in constitutive HIF-1 activation, which is associated with impaired mitochondrial biogenesis that results from HIF-1-dependent expression of MXI1, which blocks MYC-dependent expression of PGC-1β, a coactivator that is required for mitochondrial biogenesis [23]. Inhibition of wild type MYC activity in renal cell carcinoma contrasts with the synergistic effect of HIF-1 and oncogenic MYC in activating PDK1 transcription [24].

Genetic and metabolic activators of HIF-1

Hypoxia plays a critical role in cancer progression [2,5] but not all cancer cells are hypoxic and a growing number of O2-independent mechanisms have been identified by which HIF-1 is induced [5]. Several mechanisms that are particularly relevant to cancer metabolism are described below.

Activation of mTOR

Alterations in mitochondrial metabolism

NAD+ levels

It is of interest that the NAD+-dependent deacetylase sirtuin 1 (SIRT1) was found to bind to, deacetylate, and increase transcriptional activation by HIF-2α but not HIF-1α [42**]. Another NAD+-dependent enzyme is poly(ADP-ribose) polymerase 1 (PARP1), which was recently shown to bind to HIF-1α and promote transactivation through a mechanism that required the enzymatic activity of PARP1 [43]. Thus, transactivation mediated by both HIF-1α and HIF-2α can be modulated according to NAD+ levels.

Nitric oxide

Increased expression of nitric oxide (NO) synthase isoforms and increased levels of NO have been shown to increase HIF-1α protein stability in human oral squamous cell carcinoma [44]. In prostate cancer, nuclear co-localization of endothelial NO synthase, estrogen receptor β, HIF-1α, and HIF-2α was associated with aggressive disease and the proteins were found to form chromatin complexes on the promoter of TERT gene encoding telomerase [45**]. The NOS2 gene encoding inducible NO synthase is HIF-1 regulated [5], suggesting another possible feed-forward mechanism.

7.9.9 In Vivo HIF-Mediated Reductive Carboxylation

Gameiro PA1Yang JMetelo AMPérez-Carro R, et al.
Cell Metab. 2013 Mar 5; 17(3):372-85.
http://dx.doi.org/10.1016%2Fj.cmet.2013.02.002

Hypoxic and VHL-deficient cells use glutamine to generate citrate and lipids through reductive carboxylation (RC) of α-ketoglutarate. To gain insights into the role of HIF and the molecular mechanisms underlying RC, we took advantage of a panel of disease-associated VHL mutants and showed that HIF expression is necessary and sufficient for the induction of RC in human renal cell carcinoma (RCC) cells. HIF expression drastically reduced intracellular citrate levels. Feeding VHL-deficient RCC cells with acetate or citrate or knocking down PDK-1 and ACLY restored citrate levels and suppressed RC. These data suggest that HIF-induced low intracellular citrate levels promote the reductive flux by mass action to maintain lipogenesis. Using [1–13C] glutamine, we demonstrated in vivo RC activity in VHL-deficient tumors growing as xenografts in mice. Lastly, HIF rendered VHL-deficient cells sensitive to glutamine deprivation in vitro, and systemic administration of glutaminase inhibitors suppressed the growth of RCC cells as mice xenografts.

Cancer cells undergo fundamental changes in their metabolism to support rapid growth, adapt to limited nutrient resources, and compete for these supplies with surrounding normal cells. One of the metabolic hallmarks of cancer is the activation of glycolysis and lactate production even in the presence of adequate oxygen. This is termed the Warburg effect, and efforts in cancer biology have revealed some of the molecular mechanisms responsible for this phenotype (Cairns et al., 2011). More recently, 13C isotopic studies have elucidated the complementary switch of glutamine metabolism that supports efficient carbon utilization for anabolism and growth (DeBerardinis and Cheng, 2010). Acetyl-CoA is a central biosynthetic precursor for lipid synthesis, being generated from glucose-derived citrate in well-oxygenated cells (Hatzivassiliou et al., 2005). Warburg-like cells, and those exposed to hypoxia, divert glucose to lactate, raising the question of how the tricarboxylic acid (TCA) cycle is supplied with acetyl-CoA to support lipogenesis. We and others demonstrated, using 13C isotopic tracers, that cells under hypoxic conditions or defective mitochondria primarily utilize glutamine to generate citrate and lipids through reductive carboxylation (RC) of α-ketoglutarate by isocitrate dehydrogenase 1 (IDH1) or 2 (IDH2) (Filipp et al., 2012Metallo et al., 2012;Mullen et al., 2012Wise et al., 2011).

The transcription factors hypoxia inducible factors 1α and 2α (HIF-1α, HIF-2α) have been established as master regulators of the hypoxic program and tumor phenotype (Gordan and Simon, 2007Semenza, 2010). In addition to tumor-associated hypoxia, HIF can be directly activated by cancer-associated mutations. The von Hippel-Lindau (VHL) tumor suppressor is inactivated in the majority of sporadic clear-cell renal carcinomas (RCC), with VHL-deficient RCC cells exhibiting constitutive HIF-1α and/or HIF-2α activity irrespective of oxygen availability (Kim and Kaelin, 2003). Previously, we showed that VHL-deficient cells also relied on RC for lipid synthesis even under normoxia. Moreover, metabolic profiling of two isogenic clones that differ in pVHL expression (WT8 and PRC3) suggested that reintroduction of wild-type VHL can restore glucose utilization for lipogenesis (Metallo et al., 2012). The VHL tumor suppressor protein (pVHL) has been reported to have several functions other than the well-studied targeting of HIF. Specifically, it has been reported that pVHL regulates the large subunit of RNA polymerase (Pol) II (Mikhaylova et al., 2008), p53 (Roe et al., 2006), and the Wnt signaling regulator Jade-1. VHL has also been implicated in regulation of NF-κB signaling, tubulin polymerization, cilia biogenesis, and proper assembly of extracellular fibronectin (Chitalia et al., 2008Kim and Kaelin, 2003Ohh et al., 1998Thoma et al., 2007Yang et al., 2007). Hypoxia inactivates the α-ketoglutarate-dependent HIF prolyl hydroxylases, leading to stabilization of HIF. In addition to this well-established function, oxygen tension regulates a larger family of α-ketoglutarate-dependent cellular oxygenases, leading to posttranslational modification of several substrates, among which are chromatin modifiers (Melvin and Rocha, 2012). It is therefore conceivable that the effect of hypoxia on RC that was reported previously may be mediated by signaling mechanisms independent of the disruption of the pVHL-HIF interaction. Here we (1) demonstrate that HIF is necessary and sufficient for RC, (2) provide insights into the molecular mechanisms that link HIF to RC, (3) detected RC activity in vivo in human VHL-deficient RCC cells growing as tumors in nude mice, (4) provide evidence that the reductive phenotype ofVHL-deficient cells renders them sensitive to glutamine restriction in vitro, and (5) show that inhibition of glutaminase suppresses growth of VHL-deficient cells in nude mice. These observations lay the ground for metabolism-based therapeutic strategies for targeting HIF-driven tumors (such as RCC) and possibly the hypoxic compartment of solid tumors in general.

Functional Interaction between pVHL and HIF Is Necessary to Inhibit RC

Figure 1  HIF Inactivation Is Necessary for Downregulation of Reductive Carboxylation by pVHL

We observed a concurrent regulation in glucose metabolism in the different VHL mutants. Reintroduction of wild-type or type 2C pVHL mutant, which can meditate HIF-α destruction, stimulated glucose oxidation via pyruvate dehydrogenase (PDH), as determined by the degree of 13C-labeled TCA cycle metabolites (M2 enrichment) (Figures 1D and 1E). In contrast, reintroduction of an HIF nonbinding Type 2B pVHL mutant failed to stimulate glucose oxidation, resembling the phenotype observed in VHL-deficient cells (Figures 1D and 1E). Additional evidence for the overall glucose utilization was obtained from the enrichment of M3 isotopomers using [U13-C6]glucose (Figure S1A), which shows a lower contribution of glucose-derived carbons to the TCA cycle in VHL-deficient RCC cells (via pyruvate carboxylase and/or continued TCA cycling).

To test the effect of HIF activation on the overall glutamine incorporation in the TCA cycle, we labeled an isogenic pair of VHL-deficient and VHL-reconstituted UMRC2 cells with [U-13C5]glutamine, which generates M4 fumarate, M4 malate, M4 aspartate, and M4 citrate isotopomers through glutamine oxidation. As seen in Figure S1BVHL-deficient/VHL-positive UMRC2 cells exhibit similar enrichment of M4 fumarate, M4 malate, and M4 asparate (but not citrate) showing that VHL-deficient cells upregulate reductive carboxylation without compromising oxidative metabolism from glutamine. …  Labeled carbon derived from [5-13C1]glutamine can be incorporated into fatty acids exclusively through RC, and the labeled carbon cannot be transferred to palmitate through the oxidative TCA cycle (Figure 1B, red carbons). Tracer incorporation from [5-13C1]glutamine occurs in the one carbon (C1) of acetyl-CoA, which results in labeling of palmitate at M1, M2, M3, M4, M5, M6, M7, and M8 mass isotopomers. In contrast, lipogenic acetyl-CoA molecules originating from [U-13C6]glucose are fully labeled, and the labeled palmitate is represented by M2, M4, M6, M8, M10, M12, M14, and M16 mass isotopomers.

Figure 2 HIF Inactivation Is Necessary for Downregulation of Reductive Lipogenesis by pVHL

To determine the specific contribution from glucose oxidation or glutamine reduction to lipogenic acetyl-CoA, we performed isotopomer spectral analysis (ISA) of palmitate labeling patterns. ISA indicates that wild-type pVHL or pVHL L188V mutant-reconstituted UMRC2 cells relied mainly on glucose oxidation to produce lipogenic acetyl-CoA, while UMRC2 cells reconstituted with a pVHL mutant defective in HIF inactivation (Y112N or Y98N) primarily employed RC. Upon disruption of the pVHL-HIF interaction, glutamine becomes the preferred substrate for lipogenesis, supplying 70%–80% of the lipogenic acetyl-CoA (Figure 2C). This is not a cell-line-specific phenomenon, but it applies to VHL-deficient human RCC cells in general; the same changes are observed in 786-O cells reconstituted with wild-type pVHL or mutant pVHL or infected with vector only as control (Figure S2).

HIF Is Sufficient to Induce RC (reductive carboxylation) from Glutamine in RCC Cells

As shown in Figure 3C, reintroduction of wild-type VHLinto 786-O cells suppressed RC, whereas the expression of the constitutively active HIF-2α mutant was sufficient to stimulate this reaction, restoring the M1 enrichment of TCA cycle metabolites observed in VHL-deficient 786-O cells. Expression of HIF-2α P-A also led to a concomitant decrease in glucose oxidation, corroborating the metabolic alterations observed in glutamine metabolism (Figures 3D and 3E).

Figure 3 Expression of HIF-2α Is Sufficient to Induce Reductive Carboxylation and Lipogenesis from Glutamine in RCC Cells

Expression of HIF-2α P-A in 786-O cells phenocopied the loss-of-VHL with regards to glutamine reduction for lipogenesis (Figure 3G), suggesting that HIF-2α can induce the glutamine-to-lipid pathway in RCC cells per se. Although reintroduction of wild-type VHL restored glucose oxidation in UMRC2 and UMRC3 cells (Figures S3B–S3I), HIF-2α P-A expression did not measurably affect the contribution of each substrate to the TCA cycle or lipid synthesis in these RCC cells (data not shown). UMRC2 and UMRC3 cells endogenously express both HIF-1α and HIF-2α, whereas 786-O cells exclusively express HIF-2α. There is compelling evidence suggesting, at least in RCC cells, that HIF-α isoforms have overlapping—but also distinct—functions and their roles in regulating bioenergetic processes remain an area of active investigation. Overall, HIF-1α has an antiproliferative effect, and its expression in vitro leads to rapid death of RCC cells while HIF-2α promotes tumor growth (Keith et al., 2011Raval et al., 2005).

Metabolic Flux Analysis Shows Net Reversion of the IDH Flux upon HIF Activation

To determine absolute fluxes in RCC cells, we employed 13C metabolic flux analysis (MFA) as previously described (Metallo et al., 2012). Herein, we performed MFA using a combined model of [U-13C6]glucose and [1-13C1]glutamine tracer data sets from the 786-O derived isogenic clones PRC3 (VHL−/ −)/WT8 (VHL+) cells, which show a robust metabolic regulation by reintroduction of pVHL. To this end, we first determined specific glucose/glutamine consumption and lactate/glutamate secretion rates. As expected, PRC3 exhibited increased glucose consumption and lactate production when compared to WT8 counterparts (Figure 4A). While PRC3 exhibited both higher glutamine consumption and glutamate production rates than WT8 (Figure 4A), the net carbon influx was higher in PRC3 cells (Figure 4B). Importantly, the fitted data show that the flux of citrate to α-ketoglutarate was negative in PRC3 cells (Figure 4C). This indicates that the net (forward plus reverse) flux of isocitrate dehydrogenase and aconitase (IDH + ACO) is toward citrate production. The exchange flux was also higher in PRC3 than WT8 cells, whereas the PDH flux was lower in PRC3 cells. In agreement with the tracer data, these MFA results strongly suggest that the reverse IDH + ACO fluxes surpass the forward flux in VHL-deficient cells. The estimated ATP citrate lyase (ACLY) flux was also lower in PRC3 than in WT8 cells. Furthermore, the malate dehydrogenase (MDH) flux was negative, reflecting a net conversion of oxaloacetate into malate in VHL-deficient cells (Figure 4C). This indicates an increased flux through the reductive pathway downstream of IDH, ACO, and ACLY. Additionally, some TCA cycle flux estimates downstream of α-ketoglutarate were not significantly different between PRC and WT8 (Table S1). This shows that VHL-deficient cells maintain glutamine oxidation while upregulating reductive carboxylation (Figure S1B). This finding is in agreement with the higher glutamine uptake observed in VHL-deficient cells. Table S1 shows the metabolic network and complete MFA results. …

Addition of citrate in the medium, in contrast to acetate, led to an increase in the citrate-to-α-ketoglutarate ratio (Figure 5L) and absolute citrate levels (Figure S4H) not only in VHL-deficient but alsoVHL-reconstituted cells. The ability of exogenous citrate, but not acetate, to also affect RC in VHL-reconstituted cells may be explained by compartmentalization differences or by allosteric inhibition of citrate synthase (Lehninger, 2005); that is, the ability of acetate to raise the intracellular levels of citrate may be limited in (VHL-reconstituted) cells that exhibit high endogenous levels of citrate. Whatever the mechanism, the results imply that increasing the pools of intracellular citrate has a direct biochemical effect in cells with regards to their reliance on RC. Finally, we assayed the transcript and protein levels of enzymes involved in the reductive utilization of glutamine and did not observe significant differences between VHL-deficient andVHL-reconstituted UMRC2 cells (Figures S4I and S4J), suggesting that HIF does not promote RC by direct transactivation of these enzymes. The IDH1/IDH2 equilibrium is defined as follows:

[α−ketoglutrate][NADPH][CO2]/[Isocitrate][NADP+]=K(IDH)

Figure 5 Regulation of HIF-Mediated Reductive Carboxylation by Citrate Levels

We sought to investigate whether HIF could affect the driving force of the IDH reaction by also enhancing NADPH production. We did not observe a significant alteration of the NADP+/NADPH ratio between VHL-deficient and VHL-positive cells in the cell lysate (Figure S4I). Yet, we determined the ratio of the free dinucleotides using the measured ratios of suitable oxidized (α-ketoglutarate) and reduced (isocitrate/citrate) metabolites that are linked to the NADP-dependent IDH enzymes. The determined ratios (Figure S4J) are in close agreement with the values initially reported by the Krebs lab (Veech et al., 1969) and showed that HIF-expressing UMRC2 cells exhibit a higher NADP+/NADPH ratio. Collectively, these data strongly suggest that HIF-regulated citrate levels modulate the reductive flux to maintain adequate lipogenesis.

Reductive Carboxylation from Glutamine Is Detectable In Vivo

Figure 6 Evidence for Reductive Carboxylation Activity In Vivo

Loss of VHL Renders RCC Cells Sensitive to Glutamine Deprivation

We hypothesized that VHL deficiency results in cell addiction to glutamine for proliferation. We treated the isogenic clones PRC3 (VHL-deficient cells) and WT8 (VHL-reconstituted cells) with the glutaminase inhibitor 968 (Wang et al., 2010a). VHL-deficient PRC3 cells were more sensitive to treatment with 968, compared to the VHL-reconstituted WT8 cells (Figure 7A). To confirm that this is not only a cell-line-specific phenomenon, we also cultured UMRC2 cells in the presence of 968 or diluent control and showed selective sensitivity of VHL-deficient cells (Figure 7B).

Figure 7 VHL-Deficient Cells and Tumors Are Sensitive to Glutamine Deprivation

(A–E) Cell proliferation is normalized to the corresponding cell type grown in 1 mM glutamine-containing medium. Effect of treatment with glutaminase (GLS) inhibitor 968 in PRC3/WT8 (A) and UMRC2 cells (B). Rescue of GLS inhibition with dimethyl alpha-ketoglutarate (DM-Akg; 4 mM) or acetate (4 mM) in PRC3/WT8 clonal cells (C) and polyclonal 786-O cells (D). Effect of GLS inhibitor BPTES in UMRC2 cells (E). Student’s t test compares VHL-reconstituted cells to control cells in (A), (B), and (E) and DM-Akg or acetate-rescued cells to correspondent control cells treated with 968 only in (C) and (D) (asterisk in parenthesis indicates comparison between VHL-reconstituted to control cells). Error bars represent SEM.

(F) GLS inhibitor BPTES suppresses growth of human UMRC3 RCC cells as xenografts in nu/nu mice. When the tumors reached 100mm3, injections with BPTES or vehicle control were carried out daily for 14 days (n = 12). BPTES treatment decreases tumor size and mass (see insert). Student’s t test compares control to BPTES-treated mice (F). Error bars represent SEM.

(G) Diagram showing the regulation of reductive carboxylation by HIF.

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4003458/bin/nihms449661f7.jpg

In summary, our findings show that HIF is necessary and sufficient to promote RC from glutamine. By inhibiting glucose oxidation in the TCA cycle and reducing citrate levels, HIF shifts the IDH reaction toward RC to support citrate production and lipogenesis (Figure 7G). The reductive flux is active in vivo, fuels tumor growth, and can potentially be targeted pharmacologically. Understanding the significance of reductive glutamine metabolism in tumors may lead to metabolism-based therapeutic strategies.

Along with others, we reported that hypoxia and loss of VHL engage cells in reductive carboxylation (RC) from glutamine to support citrate and lipid synthesis (Filipp et al., 2012Metallo et al., 2012Wise et al., 2011). Wise et al. (2011) suggested that inactivation of HIF in VHL-deficient cells leads to reduction of RC. These observations raise the hypothesis that HIF, which is induced by hypoxia and is constitutively active inVHL-deficient cells, mediates RC. In our current work, we provide mechanistic insights that link HIF to RC. First, we demonstrate that polyclonal reconstitution of VHL in several human VHL-deficient RCC cell lines inhibits RC and restores glucose oxidation. Second, the VHL mutational analysis demonstrates that the ability of pVHL to mitigate reductive lipogenesis is mediated by HIF and is not the outcome of previously reported, HIF-independent pVHL function(s). Third, to prove our hypothesis we showed that constitutive expression of a VHL-independent HIF mutant is sufficient to phenocopy the reductive phenotype observed in VHL-deficient cells. In addition, we showed that RC is not a mere in vitro phenomenon, but it can be detected in vivo in human tumors growing as mouse xenografts. Lastly, treatment of VHL-deficient human xenografts with glutaminase inhibitors led to suppression of their growth as tumors.

7.9.10 Evaluation of HIF-1 inhibitors as anticancer agents

Semenza GL1.
Drug Discov Today. 2007 Oct; 12(19-20):853-9
http://dx.doi.org/10.1016/j.drudis.2007.08.006

Hypoxia-inducible factor 1 (HIF-1) regulates the transcription of many genes involved in key aspects of cancer biology, including immortalization, maintenance of stem cell pools, cellular dedifferentiation, genetic instability, vascularization, metabolic reprogramming, autocrine growth factor signaling, invasion/metastasis, and treatment failure. In animal models, HIF-1 overexpression is associated with increased tumor growth, vascularization, and metastasis, whereas HIF-1 loss-of-function has the opposite effect, thus validating HIF-1 as a target. In further support of this conclusion, immunohistochemical detection of HIF-1α overexpression in biopsy sections is a prognostic factor in many cancers. A growing number of novel anticancer agents have been shown to inhibit HIF-1 through a variety of molecular mechanisms. Determining which combination of drugs to administer to any given patient remains a major obstacle to improving cancer treatment outcomes.

Aurelian Udristioiu

Aurelian

Aurelian Udristioiu

Lab Director at Emergency County Hospital Targu Jiu

Mechanisms that control T cell metabolic reprogramming are now coming to light, and many of the same oncogenes importance in cancer metabolism are also crucial to drive T cell metabolic transformations, most notably Myc, hypoxia inducible factor (HIF)1a, estrogen-related receptor (ERR) a, and the mTOR pathway.
The proto-oncogenic transcription factor, Myc, is known to promote transcription of genes for the cell cycle, as well as aerobic glycolysis and glutamine metabolism. Recently, Myc has been shown to play an essential role in inducing the expression of glycolytic and glutamine metabolism genes in the initial hours of T cell activation. In a similar fashion, the transcription factor (HIF)1a can up-regulate glycolytic genes to allow cancer cells to survive under hypoxic conditions

Read Full Post »

Neutraceuticals

Author and Curator: Larry H Bernstein, MD, FCAP 

 

Introduction

This article is concerned with a very fast growing part of the food industry. Unfortunately, there have been such products that were sold that did not contain what was purported to provide the benefit. This segment of the medicinal industry is regulated as FOOD (food product), unlike pharmaceuticals. This means that the basic safety requirement is compliance with a labeling standard.  There are no clinical trials.  The incentive for the development in this sector is from 2,500 years of history in the use of plants for health benefits.  The new science that can improve the potency and the safety is the technology that has grown up to extract, refine and produce the active substance in pure form.  The methods used called Flow Chemistry has been covered recently by Anthony Melvin Crasso at his outstanding pharmaceutical site.  I shall list important articles that have been in the Pharmaceutical Intelligence site at the end of this discussion.

Antidiarrheal and antioxidant activities of chamomile (Matricaria recutita L.) decoction extract in rats

Hichem Sebai, Mohamed-Amine Jabri, Abdelaziz Souli, Kais Rtibi, et al.
J Ethnopharmacol 152(2014)327–332
http://dx.doi.org/10.1016/j.jep.2014.01.015

Ethnopharmacological relevance: Matricaria recutita L. (Chamomile) has been widely used in the Tunisian traditional medicine for the treatment of digestive system disorders. The present work aims to investigate the protective effects of chamomile decoction extract (CDE) against castor oil-induced diarrhea and oxidative stress in rats. Methods:The antidiarrheal activity was evaluated using castor oil-induced diarrhea method. In this respect, rats were divided into six groups: Control, Castor oil, Castor oil + Loperamide (LOP) and Castor oil + various doses of CDE. Animals were perorally (p.o.) pre-treated with CDE during 1 hand intoxicated for 2 or 4h by acute oral administration of castor oil.  Results: Our results showed that CDE produced a significant dose-dependent protection against castor oil-induced diarrhea and intestinal fluid accumulation. On the other hand, we showed that diarrhea was accompagnied by an oxidative stress status assessed by an increase of malondialdehyde (MDA) level and depletion of antioxidant enzyme activities as superoxide dismutase(SOD), catalase (CAT) and glutathione peroxidase (GPx). Castor oil also increased gastric and intestinal mucosa hydrogen peroxide (H2O2) and free iron levels. Importantly, we showed that chamomile pre-treatment abrogated all these biochemical alterations. Conclusion: These findings suggested that chamomile extract had a potent antidiarrheal and antioxidant properties in rats confirming their use in traditional medicine.

Anti-diarrheal activity of methanol extract of Santalum album L. in mice and gastrointestinal effect on the contraction of isolated jejunum in rats

Huimin Guo, Jingze Zhang, Wenyuan Gao, Zhuo Qu, Changxiao Liu
J Ethnopharmacol 54(2014)704–710
http://dx.doi.org/10.1016/j.jep.2014.04.043

Ethnopharmacological relevance: Santalum album L., namely Sandalwood, honoredas “Green Gold”, is a traditional Chinese herb which has the effects of antidiarrheal and antibacterial activity. But there is limited scientific study on its activity and mechanism in gastrointestinal disorders.
Materials and methods: in vivo, after intragastric administration, the methanol extract of Sandalwood (SE) (200, 400 and 800 mg/kg) were studied in castor oil-induced diarrhea mice. By the test of small intestinal hyperfunction induced by neostigmine, SE was studied on gastrointestinal transit including gastric emptying and small intestinal motility. Meanwhile, in vitro, the effects of SE (0.02, 0.05, 0.1, 0.2, 0.3, 0.4 mg/mL) on the isolated tissue preparations of rat jejunum were also investigated. The rat jejunum strips were precontacted with acetylcholine (Ach; 10-6M), 5-hydroxytryptamine (5-HT, 200 μM) or potassium chloride (KCl; 60mM) and tested in the presence of SE. In addition, the possible myogenic effect was analyzed in the pretreatment of the jejunum preparations with SE or verapamilin Ca 2+-free high-K+ (60mM) solution containing EDTA. Results: At doses of 200, 400 and 800 mg/kg, SE showed significant anti-diarrheal activity against castor oil-induced diarrhea as compared with the control. At the same doses, it also inhibited the gastric emptying and small intestinal motility in the mice of which small intestinal hyperfunction induced by neostigmine. It caused inhibitory effects on the spontaneous contraction of rat-isolated jejunum in dose-dependent manner ranging from 0.02 to 0.4 mg/mL, and it also relaxed the Ach-induced, 5-HT-induced andK+-induced contractions. SE shifted the Ca 2+concentration–response curves to right, similar to that caused by verapamil (0.025mM).
Conclusions: These findings indicated that SE played a spasmolytic role in gastrointestinal motility which was probably mediated through inhibition of muscarinic receptors, 5-HT receptors and calcium influx. All these results provide pharmacological basis for its clinical use in gastrointestinal tract.

Apocynin attenuates isoproterenol-induced myocardial injury and fibrogenesis

Li Liu, Jingang Cui, Qinbo Yang, Chenglin Jia, Minqi Xiong, et al.
Biochem Biophys Res Commun 449(2014)55–61
http://dx.doi.org/10.1016/j.bbrc.2014.04.157

Oxidative stress is mechanistically implicated in the pathogenesis of myocardial injury and the subsequent fibrogenic tissue remodeling. Therapies targeting oxidative stresss in the process of myocardial fibrogenesis are still lacking and thus remain as an active research area in myocardial injury management. The current study evaluated the effects of a NADPH oxidase inhibitor, apocynin, on the production of reactive oxygen species and  the development of myocardial fibrogenesis in isoproterenol (ISO)-induced myocardial injury mouse model. The results revealed a remarkable effect of apocynin on attenuating the development of myocardial necrotic lesions, inflammation and fibrogenesis. Additionally, the protective effects of apocynin against myocardial injuries were associated with suppressed expression of an array of genes implicated in inflammatory and fibrogenic responses. Our study thus provided for the first time the histopathological and molecular evidence supporting the therapeutic value of apocynin against the development of myocardial injuries, in particular, myocardial fibrogenesis, which will benefit the mechanism-based drug development targeting oxidative stress in preventing and/or treating related myocardial disorders.

Anti-tumor effect of Shu-gan-Liang-Xue decoction in breast cancer is related to the inhibition of aromatase and steroid sulfatase expression

Ning Zhou, Shu-Yan Han, Fei Zhou, Ping-ping Li
J Ethnopharmacol 154 (2014) 687–695
http://dx.doi.org/10.1016/j.jep.2014.04.045

Ethnopharmacological relevance: Shu-Gan-Liang-Xue Decoction (SGLXD), a traditional Chinese herbal formula used to ameliorate the hot flushes in breast cancer patients, was reported to have anti-tumor effect on breast cancer. Estrogen plays a critical role in the genesis and evolution of breast cancer. Aromatase and steroid sulfatase (STS) are key estrogen synthesis enzymes that predominantly contribute to the high local hormone concentrations. The present study was to evaluate the anti-tumor effect of SGLXD on estrogen receptor (ER) positive breast cancer celllineZR-75-1, and to investigate its underlying mechanisms both in vitro and in vivo.
Materials and methods: The anti-tumor activity of SGLXD in vitro was investigated using the MTT assay. The in vivo anti-tumor effect of SGLXD was evaluated in non-ovariectomized and ovariectomized athymic nude mice. The effect of SGLXD on enzymatic activity of aromatase and STS was examined using the dual-luciferase  reporter (DLR) based on bioluminescent measurements. Aromatase and STS protein level were assessed using Western blot assay. Results: SGLXD showed dose-dependent inhibitory effect on the proliferation of ZR-75-1 cells with IC50 value of 3.40 mg/mL. It also suppressed the stimulating effect on cell proliferation of testosterone and estrogen sulfates (E1S). Oral administration of 6 g/kg of SGLXD for 25 days resulted in a reduction in tumor volume in non-ovariectomized and ovariectomized nude mice. The bioluminescent measurements confirmed that SGLXD has a dual-inhibitory effect on the activity of aromatase and STS. Western blot assay demonstrated that the treatment of SGLXD resulted in a decrease in aromatase and STS protein levels both in vitro and in vivo. Conclusion: Our results suggested that SGLXD showed anti-tumor effect on breast cancer cells both in vitro and in vivo. The anti-tumor activity of SGLXD is related to inhibition of aromatase and STS via decreasing their expression. SGLXD may be considered as a novel treatment for ER positive breast cancer.

Cardioprotective effect of embelin on isoproterenol-induced myocardial injury in rats: Possible involvement of mitochondrial dysfunction and apoptosis

Bidya Dhar Sahu, Harika Anubolu, Meghana Koneru, et al.
Life Sciences 107 (2014) 59–67
http://dx.doi.org/10.1016/j.lfs.2014.04.035

Aims: Preventive and/or therapeutic interventions using natural products for ischemic heart disease have gained considerable attention world wide. This study investigated the cardioprotective effect and possible mechanism of embelin, a major constituent of EmbeliaribesBurm, using isoproterenol (ISO)-induced myocardial infarction model in rats.
Materials and methods: Rats were pretreated for three days with embelin (50mg/kg,p.o) before inducing myocardial injury by administration of ISO (85mg/kg) subcutaneously at aninterval of 24h for 2 consecutive days. Serum was analyzed for cardiac specific injury biomarkers, lipids and lipoprotein content. Heart tissues were isolated and were used for histopathology, antioxidant and mitochondrial respiratory enzyme activity assays and western blot analysis. Key findings: Results showed that pretreatment with embelin significantly decreased the elevated levels of serum specific cardiac injury biomarkers (CK-MB, LDH and AST), serum levels of lipids and lipoproteins and histopathological changes when compared to ISO-induced controls. Exploration of the underlying mechanisms of embelin action revealed that embelin pretreatment restored the myocardial mitochondrial respiratory enzyme activities (NADH dehydrogenase, succinate dehydrogenase, cytochrome c oxidase and mitochondrial redox activity), strengthened antioxidant status and attenuated ISO-induced myocardial lipid peroxidation. Immunoblot analysis revealed that embelin interrupted mitochondria dependent apoptotic damage by increasing the myocardial expression of Bcl-2 and downregulating the expression of Bax, cytochrome c, cleaved-caspase-3&9 and PARP. Histopathology findings further strengthened the cardioprotective findings of embelin. Significance: Result suggested that embelin may have a potential benefit in preventing ischemic heart diseasel like myocardial infarction.

Effect of Matricaria chamomilla L. flower essential oil on the growth and ultrastructure of Aspergillus niger vanTieghem

Marziyeh Tolouee, Soheil Alinezhad, Reza Saberi, Ali Eslamifar, et al.
Intl J Food Microbiol 139 (2010) 127–133
http://dx.doi.org:/10.1016/j.ijfoodmicro.2010.03.032

The antifungal activity of Matricaria chamomilla L. flower essential oil was evaluated against Aspergillus niger with the emphasis on the plant’s mode of action at the electron microscopy level. A total of 21 compounds were identified in the plant oil using gaschromatography/massspectrometry(GC/MS) accounting for 92.86% of the oil composition. The main compounds identified were α-bisabolol (56.86%), trans-trans-farnesol (15.64%), cis-β-farnesene (7.12%), guaiazulene (4.24%), α-cubebene (2.69%), α-bisabololoxideA (2.19%) and chamazulene (2.18%). In the bioassay, A.niger was cultured on Potato Dextrose Broth medium in 6-well microplates in the presence of serial two fold concentrations of plant oil (15.62 to 1000 µg/mL) for 96h at 28°C. Based on the results obtained, A. niger growth was inhibited dose dependently with a maximum of ∼92.50% at the highest oil concentration. A marked retardation in conidial production by the fungus was noticed in relation to the inhibition of hyphal growth. The main changes of hyphae observed by transmission electron microscopy were disruption of cytoplasmic membranes and intracellular organelles, detachment of plasma membrane from the cell wall, cytoplasm depletion, and completed is organization of hyphal compartments. In scanning electron microscopy, swelling and deformation of hyphal tips, formation of short branches, and collapse of entire hyphae were the major changes observed. Morphological alterations might be due to the effect on cell permeability through direct interaction of M. chamomilla essential oil with the fungal plasma membrane. These findings indicate the potential of M. chamomilla L. essential oil in preventing fungal contamination and subsequent deterioration of stored food and other susceptible materials.

Related articles in Pharmaceutical Intelligence.

Plant-based Nutrition, Neutraceuticals and Alternative Medicine: Article Compilation the Journal PharmaceuticalIntelligence.com

http://pharmaceuticalintelligence.com/2014/09/01/compilation-of-lpi-articles-on-plant-based-nutrition-and-neutraceuticals-alternative-medicine/

What do you know about Plants and Neutraceuticals?

http://pharmaceuticalintelligence.com/2014/07/30/what-do-you-know-about-plants-and-neutraceuticals/

The Discovery and Properties of Avemar – Fermented Wheat Germ Extract: Carcinogenesis Suppressor

http://pharmaceuticalintelligence.com/2014/06/07/the-discovery-and-properties-of-avemar-fermented-wheat-germ-extract-carcinogenesis-suppressor/

Introduction to Metabolomics

http://pharmaceuticalintelligence.com/2014/10/21/introduction-to-metabolomics/

Summary of Genomics and Medicine: Role in Cardiovascular Diseases

http://pharmaceuticalintelligence.com/2014/01/06/summary-of-genomics-and-medicine-role-in-cardiovascular-diseases/

Natural Products Chemistry

http://pharmaceuticalintelligence.com/2015/01/27/natural-products-chemistry/

Diet and Diabetes

http://pharmaceuticalintelligence.com/2015/03/02/diet-and-diabetes/

An Overview of Clinical Microbiology, Classification, and Antimicrobial Resistance

http://pharmaceuticalintelligence.com/2015/01/17/an-overview-of-clinical-microbiology-classification-and-antimicrobial-resistance/

Diabetes Mellitus

http://pharmaceuticalintelligence.com/2014/10/24/diabetes-mellitus/

Mechanisms of Drug Resistance

http://pharmaceuticalintelligence.com/2014/10/09/mechanisms-of-drug-resistance/

Antioxidant Potential of “Maltese Mushroom” (Cynomorium coccineum)

http://pharmaceuticalintelligence.com/2013/01/27/antioxidant-potential-of-maltese-mushroom-cynomorium-coccineum/

Advances in Separations Technology for the “OMICs” and Clarification of Therapeutic Targets

http://pharmaceuticalintelligence.com/2012/10/22/advances-in-separations-technology-for-the-omics-and-clarification-of-therapeutic-targets/

Acute Lung Injury

http://pharmaceuticalintelligence.com/2015/02/26/acute-lung-injury/

The Vibrant Philly Biotech Scene: Focus on KannaLife Sciences and the Discipline and Potential of Pharmacognosy

Curator and Interviewer: Stephen J. Williams, Ph.D.

http://pharmaceuticalintelligence.com/2015/02/19/the-vibrant-philly-biotech-scene-focus-on-kannalife-sciences-and-the-discipline-and-potential-of-pharmacognosy/

Complex Models of Signaling: Therapeutic Implications

http://pharmaceuticalintelligence.com/2014/10/31/complex-models-of-signaling-therapeutic-implications/

Parasitic Plant Strangleweed Injects Host With Over 9,000 RNA Transcripts

Reporter: Aviva Lev-Ari, PhD, RN

http://pharmaceuticalintelligence.com/2014/09/02/parasitic-plant-strangleweed-injects-host-with-over-9000-rna-transcripts/

Health benefit of anthocyanins from apples and berries noted for men

http://pharmaceuticalintelligence.com/2014/07/06/health-benefit-of-anthocyanins-from-apples-and-berries-noted-for-men/

Requiem for Palliative Cardiology: The Voice of Dr. Esselstyn on Plant-Based Nutrition

Reporter: Aviva Lev-Ari, PhD, RN

http://pharmaceuticalintelligence.com/2014/02/02/requiem-for-palliative-cardiology-the-voice-of-dr-esselstyn-on-plant-based-nutrition/

Read Full Post »

Stress and Anxiety

Writer and Curator: Larry H Bernstein, MD, FCAP

 

Introduction

This article follows immediately after two on diet and obesity and diet and exercise. The hypothalamus has been discussed in some detail, although There is more that needs to be said about glutamate receptors, which is a topic in itself. However, this material fits in place quite well.  There is a considerable amount of obesity, and exercise is limited by time and commitment.  The shrinking middle class and the working poor, and the unemployed poor as well, have a struggle to make ends meet, and with the divorce rates that we are seeing, it is stressful for a single mother to carry on a complete life as mother and caregiver, and it is not unusual to see one or both couples in a household, regardless of sex, to hold two jobs.  Students enter colleges for higher education and leave with significant debts.  Graduates with advanced degrees may have to compete with a crowd of qualified applicants for an academic position, or even for a job in technology.  In addition, there is an increase in stress related disorders in the   pre-school, elementary and middle school population.  We no longer have to read the front pages to learn that a violent act has been carried out somewhere, in some neighborhood in our great nation that has experienced a great civil war, two world wars, the Mc Carthy hearings, the Cold War, and Vietnam, and the Iraq War, all of which was accompanied by migrations, immigration, and outsourcing of jobs.  The following is another look at how we are adjusting.

 

Effectiveness of a meditation-based stress management program as an adjunct to pharmacotherapy in patients with anxiety disorder

Sang Hyuk Lee, Seung Chan Ahn, Yu Jin Lee, Tae Kyu Choi, et al.
J Psychosomatic Research 62 (2007) 189–195
http://dx.doi.org:/10.1016/j.jpsychores.2006.09.009

Objective: The objective of this study was to examine the effectiveness of a meditation-based stress management program in patients with anxiety disorder.
Methods: Patients with anxiety disorder were randomly assigned to an 8-week clinical trial of either a meditation-based stress management program or an anxiety disorder education program. The Hamilton Anxiety Rating Scale (HAM-A), the Hamilton Depression Rating Scale (HAM-D), the State–Trait Anxiety Inventory (STAI), the Beck Depression Inventory, and the Symptom Checklist- 90 — Revised (SCL-90-R) were used to measure outcome at 0, 2, 4, and 8 weeks of the program. Results: Compared to the education group, the meditation-based stress management group showed significant improvement in scores on all anxiety scales (HAM-A, P=.001; STAI state, P=.001; STAI trait, P=.001; anxiety subscale of SCL-90-R,P=.001) and in the SCL-90-R hostility subscale (P=.01). Findings on depression measures were inconsistent, with no significant improvement shown by subjects in the meditation-based stress management group compared to those in the education group. The meditation-based stress management group did not show significant improvement in somatization, obsessive–compulsive symptoms, and interpersonal sensitivity scores, or in the SCL-90-R phobic anxiety subscale compared to the education group. Conclusions: A meditation-based stress management program can be effective in relieving anxiety symptoms in patients with anxiety disorder. However, well-designed, randomized, and controlled trials are needed to scientifically prove the worth of this intervention prior to treatment.

 

Evidence and Potential Mechanisms for Mindfulness Practices and Energy Psychology for Obesity and Binge-Eating Disorder

Renee Sojcher, Susan Gould Fogerite, and Adam Perlman
Explore 2012; 8(5):271-276
http://dx.doi.org/10.1016/j.explore.2012.06.003

Obesity is a growing epidemic. Chronic stress produces endocrine and immune factors that are contributors to obesity’s etiology. These biochemical alsocan affect appetite and eating behaviors that can lead to binge-eating disorder. The inadequacies of standard care and the problem of patient noncompliance have inspired a search for alternative treatments. Proposals in the literature have called for combination therapies involving behavioral or new biological therapies. This manuscript suggests that mindbody interventions would be ideal for such combinations. Two mind body modalities, energy psychology and mindfulness meditation, are reviewed for their potential in treating weight loss, stress, and behavior modification related to binge-eating disorder.

Whereas mindfulness meditation and practices show more compelling evidence, energy psychology, in the infancy stages of elucidation, exhibits initially promising outcomes but requires further evidence-based trials. “Diets Don’t Work” has been a mantra repeated over and over in the media. In fact, in a 2006 study in which investigators compared several popular diets comprising either high carbohydrates, high protein, or high fat, they found a rapid regression of compliance after six months, to the extent that it did not matter which diet had initially been more effective. In another study, authors examined a combination of diet and exercise compared with diet alone and observed that 50% of their subjects in both groups regained the weight that they lost after one year, despite their having lost more weight with the combination therapy. Despite the failure of diet alone in most studies, strategies incorporating both diet and exercise can be effective: a Cochrane review on exercise for overweight or obesity concluded that exercise had a positive effect on body weight and cardiovascular risk factors and that this effect was enhanced by a combination of exercise with dietary interventions.

The authors of a more recent study found that the benefits of exercise in inducing weight loss may come through psychological pathways rather than through actual energy expenditure. These factors include self-regulation and self-efficacy, which may mediate the relationship between exercise and weight change. Psychological interventions, particularly behavioral therapy and CBT, have been shown to be effective, especially when combined with diet and exercise. However, these interventions are costly and require extensive clinical contact for long durations to achieve efficacy. The authors of a recent randomized controlled trial (RCT) with a three-year follow-up period looked at a new form of CBT that addresses patients’ overeating and low level of activity, as well as factors that impede weight maintenance, and found that this form of therapy did not result in improved weight maintenance. These authors concluded that CBT is not sufficiently effective in helping patients maintain their weight loss in the long term. Although 20% of people will not change their eating behaviors under stress, most do; approximately 40% will increase and 40% will decrease their eating.

The emotional eaters, who tend to increase food intake, are more likely to crave high-fat/sweet and rewarding comfort foods. The basis for this behavior is becoming understood to entail brain pathways that involve learning and memory of reward and pleasure. Habit formation and decreased cognitive control are also involved. These habits form the basis of BED. Binge eating occurs when a person eats larger amounts of food than normal in a short amount of time. It therefore involves a loss of control and is often precipitated by a range of negative emotions, such as anxiety, depression, anger, and loneliness. Overweight subjects may or may not be characterized as binge eaters.

The stress response, also known as the “fight or flight response,” involves the interaction of the autonomic nervous system, which includes the sympathetic and the parasympathetic nervous systems, the hypothalamic pituitary adrenal axis and endocrine secretion. Together, these systems comprise neuro-endocrine pathways that collaborate to maintain the body’s regulation of homeostasis. This mechanism is very effective when stress is acute, but in the case of chronic stress, the effect can be injurious to one’s physiological state. Over time, chronic exposure to stress hormones contributes to“ allostatic load.” The stress hormones released by the body, mostly cortisol, can alter the body’s fuel metabolism, especially by adipose tissue, leading to an increase in upper-body obesity. Furthermore, hormones such as leptin, ghrelin, and neuropeptide Y can affect appetite and cause changes in fat mass storage. This results in the linking of stress and obesity.

Given the limited success of conventional approaches and the new information about the psychological and physiological mechanisms underlying obesity, we propose that a specific sub-group of mind-body therapies, including energy psychology and mindfulness-based approaches, could add an important new dimension to the integrative treatment of eating disorders. Energy psychology refers to a family of therapies that are used for treating physical disorders and psychological symptoms, which includes Thought Field Therapy, Emotional Freedom Techniques (EFT), Eye Movement Desensitization and Reprocessing, and Tapas Acupressure Technique (TAT). These therapies incorporate concepts originating from non-Western healing and spiritual systems, including acupuncture, acupressure, yoga, meditation, and qigong, and they combine physical activity with mental activation on the basis of the premise that the body is composed of electrical signals or energy fields. Energy psychology has been quite controversial among psychotherapists and has been the subject of much heated debate in the literature. Nonetheless, the clinical application of these practices is growing and is beginning to be investigated for efficacy. Mindfulness-Based Eating Awareness Training (ie,MB-EAT) involves the cultivation of mindfulness, mindful eating, emotional balance, and self-acceptance.

A pilot trial of a six-week group curriculum for providing mindfulness training to obese individuals, called Mindful Eating and Living (ie,MEAL), showed significant increases in measures of mindfulness and cognitive restraint around eating and significant decreases in weight, eating disinhibition, bingeeating, depression, perceived stress, physical symptoms, negative affect ,and C-reactive protein. In a recent systematic review of eight studies, authors examined a variety of mindfulness techniques in treating eating disorders, including anorexia, bulimia, and BED. Because trial quality varied and sample sizes were small, the researchers concluded that mindfulness may be effective in treating eating disorders but that further research was needed. The authors noted, however, that all of the articles that met the study’s criterion reported positive outcomes for the mindfulness intervention. Two additional studies recently addressed the treatment of obesity with a combination of mindfulness strategies and ACT. Lillis et al. conducted a RCT on 87 subjects who had all completed at least a six-month weight loss program. Using a wait list control against treatment of the experimental group through a one-day workshop, the authors found that, compared with the control group, the experimental group showed greater improvements in obesity-related stigma, quality of life, psychological distress, and reduction of body mass in a three-month follow-up. Alberts et al. conducted an RCT on 19 participants in a 10-week dietary group treatment that examined the effect of mindfulness plus ACT on food cravings. Experimental subjects underwent an additional seven-week, manual-based mindfulness/acceptance training. The control group received information on healthy food choices. The experimental group showed significantly lower food cravings, a lower preoccupation with food in four subscales, less loss of control, and better positive outcome expectancy, as compared with the control group. There was no significant effect observed for emotional craving. The authors of both of these studies conclude that mindfulness strategies combined with acceptance are effective in reducing the behaviors that lead many obese patients to overeat. With regards to stress, mindfulness can reduce psychological factors that have been shown to contribute to obesity.

In a recent well conducted systematic review, Mars and Abbey examined 22 studies with conditions ranging from participants with Axis I disorders, various diagnosed medical disorders, and healthy subjects. Axis I disorders include a range of psychopathologies such as childhood developmental and adjustment abnormalities, adult anxiety, and mood, sleep, and sexual disorders. Subjects with BED are known to have greater comorbidity forAxis I disorders. The authors report that five studies examining Axis I disorders showed statistically significant results for an eight-week, two hours per week MBCT program in reducing psychological stress, recurring bouts of depression, and pain. They conclude that, despite some methodological difficulties in the trials, mindfulness therapy may have a positive impact on reducing stress and depression. Despite increasing public awareness of obesity’s detrimental effects on health, the conventional approaches to managing this condition have not been effective. The recommended standard care for overweight and obesity, namely diet and exercise, are for the most part ineffective in the long term. Behavioral therapy and CBT may have some effect but are costly and difficult to implement. Issues with bariatric surgery and pharmacological therapies attributable to cost and the potential for harm, as well as lack of long-term efficacy, have limited their utility.

The effectiveness of a stress coping program based on mindfulness meditation on the stress, anxiety, and depression experienced by nursing students in Korea

Yune Sik Kang, So Young Choi, Eunjung Ryu
Nurse Education Today 29 (2009) 538–543
http://dx.doi.org:/10.1016/j.nedt.2008.12.003

This study examined the effectiveness of a stress coping program based on mindfulness meditation on the stress, anxiety, and depression experienced by nursing students in Korea. A nonequivalent, control group, pre-posttest design was used. A convenience sample of 41 nursing students were randomly assigned to experimental (n=21) and control groups (n=20). Stress was measured with the PWI-SF(5-point) developed by Chang. Anxiety was measured with Spieberger’s state anxiety y inventory. Depression was measured with the Beck depression inventory. The experimental group attended 90-min sessions for eight weeks. No intervention was administered to the control group. Nine participants were excluded from the analysis because they did not complete the study due to personal circumstances, resulting in16 participants in each group for the final analysis. Results for the two groups showed

(1) a significant difference in stress scores (F=6.145,p=0.020),

(2) a significant difference in anxiety scores (F=6.985,p=0.013), and

(3) no significant difference in depression scores (t=1.986,p=0.056).

A stress coping program based on mindfulness meditation was an effective intervention for nursing students to decrease their stress and anxiety, and could be used to manage stress in student nurses. In the future, long-term studies should be pursued to standardize and detail the program, with particular emphasis on studies to confirm the effects of the program in patients with diseases, such as cancer.

 

 

Meditation and Anxiety Reduction: A Literature Review

M. M. Delmonte Clin
Psychol Rev 1985; 5: 91-102
Meditation is increasingly being practiced as a therapeutic technique. The effects of practice on psychometrically assessed anxiety levels has been extensively researched. Prospective meditators tend to report above average anxiety. In general, high anxiety levels predict a subsequent low frequency of practice. However, the evidence suggests that those who practice regularly tend to show significant decreases in anxiety. Meditation does not appear to be more effective than comparative interventions in reducing anxiety. There is evidence to suggest that hypnotizability and expectancy may both play a role in reported anxiety decrease. Certain individuals with a capacity to engage in autonomous self-absorbed relaxation, may benefit most from meditation.

 

Meta-analysis on the effectiveness of mindfulness-based stress reduction therapy on mental health of adults with a chronic disease: What should the reader not make of it?

Ernst Bohlmeijer, Rilana Prenger, ErikTaal
Letters to the Editor/J Psychosom Res 69 (2010) 613–615
http://dx.doi.org:/10.1016/j.jpsychores.2010.09.005

In a letter to the editor, Nyklíček et al. discuss the study of Bohlmeijer et al. [1]on the meta-analysis on the effectiveness of mindfulness-based stress reduction (MBSR) therapy on mental health of adults with a chronic disease. They claim that the effects of MBSR are underestimated in this meta-analysis due to the inclusion of a study using an active education support group as control group and to the omission of some subscales for which larger effect sizes have been found. We do not agree that the study using an active education support group as a control group should not have been included in the meta-analysis. It is a common procedure to include studies with various types of control groups, e.g., waiting-list, placebo, minimal interventions, or evidence-based treatment. Normally, subgroup analyses can be conducted, contrasting studies that use differen ttypes of control groups. As seven studies used a waiting-list control condition and only one study used an education support group, this subgroup comparison was not useful. However, when we conducted a meta-analysis of the seven RCTs using a waiting-list control group an overall effect size of 0.30 instead of 0.26 was found. In addition, it is often found in meta-analyses that the largest effect sizes are reported in studies that use waiting-list control groups, e.g. ,Refs.[2,3]. The fact that almost all studies included in our meta-analysis in fact used waiting-list control groups makes it unlikely that the effects of MBSR were underestimated. As to the second claim by Nyklíček e tal.that some outcomes were selectively omitted from the meta-analysis, we can state that the subscales of the POMS were included in the meta-analysis.The program that was used in our study, Comprehensive Meta-Analysis, combined the scales that measure the same outcome, e.g., anxiety in one study. So the larger effects sizes for the subscales of the POMS were included in the meta-analysis. Lastly, Nyklíčeketal. State that ‘decentering’ is not an exclusive process of MBCT but is a central feature of MBSR as well. MBCT was specifically developed for people with recurrent depression and on the basis of a thorough analysis of the role of specific cognitions in people with recurrent depression. In ouropinion, this may explain the large effect sizes that have been found in randomized controlled trials, e.g., [4]. In general, other studies have shown that integrating MBSR in behavioral therapy is a very promising strategy for enhancing the efficacy of treatments of psychological  distress[5,6]. However, more studies with different target groups are needed to answer the question as to which mindfulness-based intervention is most effective for which target group in which setting. Overall, in response to the letter to the editor by Nyklíček et al. we cannot corroborate their claim that the effects of MBSR were underestimated and have to stand with our conclusion that, on the basis of current RCTs, MBSR has small leffects on depression and anxiety in people with chronic medical diseases.

[1] BohlmeijerET, PrengerR, TaalE, CuijpersP.
The effects of mindfulness-based stress reduction therapy on the mental health of adults with a chronic medical disease: A meta-analysis.
JPsychosom Res 2010; 68:539–44.

[2]Powers MB, Zum Vörde Sive Vörding MB, Emmelkamp PMG.
Acceptance and commitment therapy: A meta-analytic review.
Psychoth Psychosom 2009; 78:73–80.

Read Full Post »

Diet and Cholesterol

Writer and Curator: Larry H. Bernstein, MD, FCAP 

 

Introduction

We are all familiar with the conundrum of diet and cholesterol.  As previously described, cholesterol is made by the liver. It is the backbone for the synthesis of sex hormones, corticosteroids, bile, and vitamin D. It is also under regulatory control, and that is not fully worked out, but it has health consequences. The liver is a synthetic organ that is involved with glycolysis, gluconeogenesis, cholesterol synthesis, and unlike the heart and skeletal muscles – which are energy transducers – the liver is anabolic, largely dependent on NADPH.  The mitochondria, which are associated with aerobic metabolism, respiration, are also rich in the liver.  The other part of this story is the utilization of lipids synthesized by the liver in the vascular endothelium.  The vascular endothelium takes up and utilizes/transforms cholesterol, which is involved in the degenerative development of pathogenic plaque.  Plaque is associated with vascular rigidity, rupture and hemorrhage, essential in myocardial inmfarction. What about steroid hormones?  There is some evidence that sex hormone differences may be a factor in coronary vascular disease and cardiac dysfunction.  The evidence that exercise is beneficial is well established, but acute coronary events can occur during exercise.  WE need food, and food is at the center of the discussion – diet and cholesterol.  The utilization of food varies regionally, and is dependent on habitat.  But it is also strongly influence by culture.  We explore this further in what follows.

A high fat, high cholesterol diet leads to changes in metabolite patterns in pigs – A metabolomic study

Jianghao Sun, Maria Monagas, Saebyeol Jang, Aleksey Molokin, et al.
Food Chemistry 173 (2015) 171–178
http://dx.doi.org/10.1016/j.foodchem.2014.09.161

Non-targeted metabolite profiling can identify biological markers of dietary exposure that lead to a better understanding of interactions between diet and health. In this study, pigs were used as an animal model to discover changes in metabolic profiles between regular basal and high fat/high cholesterol diets. Extracts of plasma, fecal and urine samples from pigs fed high fat or basal regular diets for 11 weeks were analysed using ultra-high performance liquid chromatography with high-resolution mass spectrometry (UHPLC–HRMS) and chemometric analysis. Cloud plots from XCMS online were used for class separation of the most discriminatory metabolites. The major metabolites contributing to the discrimination were identified as bile acids (BAs), lipid metabolites, fatty acids, amino acids and phosphatidic acid (PAs), phosphatidylglycerol (PGs), glycerophospholipids (PI), phosphatidylcholines (PCs) and tripeptides. These results suggest the developed approach can be used to identify biomarkers associated with specific feeding diets and possible metabolic disorders related to diet.

Nutritional metabolomics is a rapidly developing sub-branch of metabolomics, used to profile small-molecules to support integration of diet and nutrition in complex bio-systems research. Recently, the concept of ‘‘food metabolome’’ was introduced and defined as all metabolites derived from food products. Chemical components in foods are absorbed either directly or after digestion, undergo extensive metabolic modification in the gastrointestinal tract and liver and then appear in the urine and feces as final metabolic products. It is well known that diet has a close relationship with the long-term health and well-being of individuals. Hence, investigation of the ‘‘food metabolome’’ in biological samples, after feeding specific diets, has the potential to give objective information about the short- and long-term dietary intake of individuals, and to identify potential biomarkers of certain dietary patterns. Previous studies have identified potential biomarkers after consumption of specific fruits, vegetables, cocoa, and juices. More metabolites were revealed by using metabolomic approaches compared with the detection of pre-defined chemicals found in those foods.

Eating a high-fat and high cholesterol diet is strongly associated with conditions of obesity, diabetes and metabolic syndrome, that are increasingly recognized as worldwide health concerns. For example, a high fat diet is a major risk factor for childhood obesity, cardiovascular diseases and hyperlipidemia. Little is known on the extent to which changes in nutrient content of the human diet elicit changes in metabolic profiles. There are several reports of metabolomic profiling studies on plasma, serum, urine and liver from high fat-diet induced obese mice, rats and humans. Several potential biomarkers of obesity and related diseases, including lysophosphatidylcholines (lysoPCs), fatty acids and branched-amino acids (BCAAs) have been reported.

To model the metabolite response to diet in humans, pigs were fed a high fat diet for 11 weeks and the metabolite profiles in plasma, urine and feces were analyzed. Non-targeted ultra high performance liquid chromatography tandem with high resolution mass spectrometry (UHPLC–MS) was utilized for metabolomics profiling. Bile acids (BAs), lipid metabolites, fatty acids, amino acids and phosphatidic acid (PAs), phosphatidylglycerol (PGs), glycerophospholipids (PI), phosphatidylcholines (PCs), tripeptides and isoflavone conjugates were found to be the final dietary metabolites that differentiated pigs fed a high-fat and high cholesterol diet versus a basal diet. The results of this study illustrate the capacity of this metabolomic profiling approach to identify new metabolites and to recognize different metabolic patterns associated with diet.

Body weight, cholesterol and triglycerides were measured for all the pigs studied. There was no significant body weight gain between pigs fed diet A and diet B after 11 weeks of treatment. The serum cholesterol and triglyceride levels were significantly higher in pigs fed with diet B compared with the control group at the end of experiment.

Plasma, urine and fecal samples were analyzed in both positive and negative ionization mode. To obtain reliable and high-quality metabolomic data, a pooled sample was used as a quality control (QC) sample to monitor the run. The QC sample (a composite of equal volume from 10 real samples) was processed as real samples and placed in the sample queue to monitor the stability of the system. All the samples were submitted in random for analysis. The quantitative variation of the ion features across the QC samples was less than 15%. The ion features from each possible metabolite were annotated by XCMS online to confirm the possible fragment ions, isotopic ions and possible adduct ions. The reproducibility of the chromatography was determined by the retention time variation profiles that were generated by XCMS. The retention time deviation was less than 0.3 min for plasma samples, less than 0.3 min for fecal samples, and less than 0.2 min for urine samples, respectively. On the basis of these results of data quality assessment, the differences between the test samples from different pigs proved more likely to reflect varied metabolite profiles rather than analytical variation. The multivariate analysis results from the QC sample showed the deviation of the analytical system was acceptable.
Good separation can be observed between pigs on the two diets, which is also reflected in the goodness of prediction (Q2), of 0.64 using data from the positive ionization mode. For negative ionization mode data, better separation appears with a Q2of 0.73.

Cloud plot is a new multidimensional data visualization method for global metabolomic data (Patti et al., 2013). Data characteristics, such as the p-value, fold change, retention time, mass-to-charge ratio and signal intensity of features, can be presented simultaneously using the cloud plot. In this study, the cloud plot was used to illustrate the ion features causing the group separation. In Fig. 2 and 82 features with p < 0.05 and fold change >2, including visualisation of the p-value, the directional fold change, the retention time and the mass to charge ratio of features, are shown. Also, the total ion chromato-grams for each sample were shown. The upper panel in (2A) shows the chromatograms of plasma samples from pigs fed the high fat diet, while the lower panel shows the chromatograms of samples from pigs fed the regular diet. Features whose intensity is increased are shown in green, whereas features whose intensity is decreased are shown in pink (2A). The size of each bubble corresponds to the log fold change of the feature: the larger the bubble, the larger the fold changes. The statistical significance of the fold change, as calculated by a Welch t-test with unequal variances, is represented by the intensity of the feature’s color where features with low p-values are brighter compared to features with high p-values. The Y coordinate for each feature corresponds to the mass-to-charge ratio of the compound, as determined by mass spectrometry. Each feature is also color coded, such as features that are shown with a black outline have database hits in METLIN, whereas features shown without a black outline do not have any database hits.

From the cloud plot (Fig. 2A), 82 discriminating ion features from positive data and 48 discriminating ions features from negative data were considered as of great importance for class separation. After filtering out the fragment ions, isotope annotations, and adduct ions, thirty-one metabolites were tentatively assigned using a Metlin library search (Table S4).

Among the assigned metabolites detected, five of the highest abundant metabolites were identified as bile acid and bile acid conjugates (Fig. 2B). This series of compounds shared the following characteristics; the unconjugated bile acids showed [M-H] ion as base peak in the negative mode.

The characteristic consistent with bile acid hyodeoxycholic acid (HDCA) was confirmed with a reference standard. For the conjugated bile acids (usually with glycine and taurine), the [M-H] and [M+H]+ are always observed as the base peaks. For example, the ion feature m/z 448.3065 at 21.18 min was identified as chenodeoxycholic acid glycine conjugate. The neutral loss of 62 amu (H2O + CO2) was considered as a characteristic fragmentation pathway for bile acid glycine conjugates. This above mentioned characteristic can easily identify a series of bile acids compounds. The five metabolite ions detected in plasma were significantly different between pigs fed the high fat diet (Fig. 2B, red bars) and regular diet (Fig. 2B, blue bars) for 11 weeks, and were identified as chenodeoxycholic acid glycine conjugate, tauroursodeoxycholic acid, hyodeoxycholic acid, deoxycholic acid glycine conjugate and glycocholic acid; chenodeoxycholic acid glycine and hyodeoxycholic acid.

Figures 1-4 , not shown.
Fig 1. The PCA score plot of plasma (A) (+)ESI data with all the ion features; (B) (+)ESI data with selected ion features; (C) (-)ESI data with all ion features; (D) (-)ESI data with selected ion features. Samples were taken from pigs fed diet A (BS, blue) and diet B (HF, red). (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

Fig 2. Cloud plot showing 82 discriminatory ion features (negative ion data) in plasma, and (B) box-plot of data set of the five most abundant bile acids identified in plasma (negative ion data) samples.

Fig. 3. PCA score plot of fecal samples from pigs fed diet A (BS, blue) and diet B (HF, red) (A) week 0, (B) week 2, (C) week 4 (D) week 6, (E) week 11 for distal samples (F) week 11 for proximal colon samples. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

Fig. 4. PCA and PLS-DA score plot of urine samples from (+)ESI-data (A and C) and (-)ESI-data (B and D) taken at the end of the study (week 11) from pigs fed diet A (BS, blue) and diet B (HF, red). (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

Plasma, fecal and urine metabolites from pigs fed either a high fat or regular diet were investigated using a UHPLC–HRMS based metabolomic approach. Their metabolic profiles were compared by multivariate statistical analysis.
Diet is logically believed to have a close relationship with metabolic profiles. Feeding a high fat and high cholesterol diet to pigs for 11 weeks resulted in
an increase in bile acids and their derivatives in plasma, fecal and urine samples, though at this stage, there was no significant weight gain observed.

In a previous study, a significantly higher level of muricholic acid, but not cholic acid, was found in pigs fed a high fat diet. The gut microbiota of these pigs were altered by diet and considered to regulate bile acid metabolism by reducing the levels of tauro-beta-muricholic acid. In our study, the unconjugated bile acids, hyodeoxycholic acid and deoxycholic acid were found to be significantly higher in the fecal samples of pigs fed a high-fat diet.

Chenodeoxycholic acid glycine was 8.6 times higher in pigs fed a high fat and high cholesterol diet compared to those fed a regular diet. These results confirm that feeding a high fat and high cholesterol diet leads to a changing metabolomic pattern over time, represented by excretion of certain bile acids in the feces. We also found that several metabolites associated with lipid metabolism were increased in the feces of pigs fed the high-fat diet. Feeding the high fat diet to pigs for 11 weeks did not induce any overt expression of disease, except for significantly higher levels of circulating cholesterol and triglycerides in the blood. It is likely, however, that longer periods of feeding would increase expression of metabolic syndrome disorders and features of cardiovascular disease in pigs, as have been previously demonstrated. Products of lipid metabolism that changed early in the dietary treatment could be useful as biomarkers. This may be important because the composition of the fats in the diet, used in this study, was complex and from multiple sources including lard, soybean oil and coconut oil.

In summary, a number of metabolite differences were detected in the plasma, urine and feces of pigs fed a high fat and high cholesterol diet versus a regular diet that significantly increased over time. PCA showed a clear separation of metabolites in all biological samples tested from pigs fed the different diets. This methodology could be used to associate metabolic profiles with early markers of disease expression or the responsiveness of metabolic profiles to alterations in the diet. The ability to identify metabolites from bio-fluids, feces, and tissues that change with alterations in the diet has the potential to identify new biomarkers and to better understand mechanisms related to diet and health.

Amino acid, mineral, and polyphenolic profiles of black vinegar, and its lipid lowering and antioxidant effects in vivo

Chung-Hsi Chou, Cheng-Wei Liu, Deng-Jye Yang, Yi-Hsieng S Wuf, Yi-Chen Chen
Food Chemistry 168 (2015) 63–69
http://dx.doi.org/10.1016/j.foodchem.2014.07.035

Black vinegar (BV) contains abundant essential and hydrophobic amino acids, and polyphenolic contents, especially catechin and chlorogenic acid via chemical analyses. K and Mg are the major minerals in BV, and Ca, Fe, Mn, and Se are also measured. After a 9-week experiment, high-fat/cholesterol-diet (HFCD) fed hamsters had higher (p < 0.05) weight gains, relative visceral-fat sizes, serum/liver lipids, and serum cardiac indices than low-fat/cholesterol diet (LFCD) fed ones, but BV supplementation decreased (p < 0.05) them which may resulted from the higher (p < 0.05) fecal TAG and TC contents. Serum ALT value, and hepatic thiobarbituric acid reactive substances (TBARS), and hepatic TNF-α and IL-1β contents in HFCD-fed hamsters were reduced (p < 0.05) by supplementing BV due to increased (p < 0.05) hepatic glutathione (GSH) and trolox equivalent antioxidant capacity (TEAC) levels, and catalase (CAT) and glutathione peroxidase (GPx) activities. Taken together, the component profiles of BV contributed the lipid lowering and antioxidant effects on HFCD fed hamsters.

World Health Organization (WHO) reported that more than 1.4 billion adults were overweight (WHO, 2013). As we know, imbalanced fat or excess energy intake is one of the most important environmental factors resulted in not only increased serum/liver lipids but also oxidative stress, further leading cardiovascular disorders and inflammatory responses. Food scientists strive to improve serum lipid profile and increase serum antioxidant capacity via  medical foods or functional supplementation.

Vinegar is not only used as an acidic seasoning but also is shown to have some beneficial effects, such as digestive, appetite stimulation, antioxidant, exhaustion recovering effects, lipid lowering effects, and regulations of blood pressure. Polyphenols exist in several food categories, such as vegetable, fruits, tea, wine, juice, and vinegar that have effects against lipid peroxidation, hypertension, hyperlipidemia, inflammation, DNA damage, and. Black vinegar (BV) (Kurosu) is produced from unpolished rice with rice germ and bran through a stationary surface fermentation and contains higher amounts of amino acids and organic acids than other vinegars. Black vinegar is also characterised as a health food rather than only an acidic seasoning because it was reported to own a DPPH radical scavenging ability and decrease the adipocyte size in rat models. Moreover, the extract of BV shows the highest radical scavenging activity in a DPPH radical system than rice, grain, apple, and wine vinegars. The extract suppresses increased lipid peroxidation in mouse skin treated with 12-o-tetradecanoylphorbol-13-acetate.

This study focused on the nutritional compositions in BV, and the in-vivo lipid lowering and antioxidant effects. First, the amino acid, mineral, and polyphenolic profile of BV were identified. Hypolipidemic hamsters induced by a high-fat/cholesterol diet (HFCD) were orally administered with different doses of BV. Serum lipid profile and liver damage indices liver and fecal lipid contents, as well as hepatic antioxidant capacities [thiobarbituric acid reactive substances (TBARS), glutathione (GSH), trolox equivalent antioxidant capacity (TEAC), and activities of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx)] and hepatic cytokine levels were assayed to demonstrated physiological functions of BV.

Higher serum AST, ALT, and free fatty acids, as well as hepatic cholesterol, triacylglycerol, MDA, hydroperoxide, and cytokine (IL-1β and TNF-α) levels were easily observed in a high-fat-consumption rodent. Several reports indicated some amino acids antioxidant activities in vitro and in vivo. Acidic amino acids, such as Asp and Glu and hydrophobic amino acids, such as Ile, Leu, and Val display high antioxidant properties. Recently, an in vivo study indicated that a pepsin hydrolyzation significantly enhanced Asp, Glu, Leu, and Val contents in chicken livers; meanwhile, chicken-liver hydrolysates showed an antioxidant capacity in brain and liver of D-galactose treated mice. In addition, it was also reported that Mg and Se play important roles in SOD and GPx activities, respectively. Uzun and Kalender (2013) used chlorpyrifos, an organophosphorus insecticide, to induce hepatotoxic and hematologic changes in rats, but they observed that catechin can attenuate the chlorpyrifos-induced hepatotoxicity by increasing GPx and glutathione-S-transferase activities and decreasing MDA contents. Meanwhile, chlorogenic acid elevated SOD, CAT, and GPx activities with concomitantly decreased lipid peroxidation of liver and kidney in streptozotocin-nicotinamide induced type-2 diabetic rats. Hence, it is reasonable to assume that increased antioxidant capacities and decreased damage in livers of HFCD fed hamsters supplemented with BV should be highly related to the components, i.e. amino acid profile, mineral profile, and polyphenol contents, as well as the lowered liver lipid accumulations.

In analyses of amino acids, minerals and polyphenols, BV contained abundant essential amino acids and hydrophobic amino acids. Mg, K, Ca, Fe, Mn, and Se were measured in BV where K and Mg were major. Gallic acid, catechin, chlorogenic acid, p-hydroxybezoic acid, p-cumeric acid, ferulic acid, and sinapic acid were also identified in BV where catechin and chlorogenic acid were the majorities. Meanwhile, the lipid-lowering and antioxidant effects of BV were also investigated via a hamster model. BV supplementation apparently decreased weight gain (g and %), relative size of visceral fat, serum/liver TC levels, serum cardiac index, and hepatic TBARS values and damage indices (serum ALT and hepatic TNF-α and IL-1β) but increased fecal lipid contents and hepatic antioxidant capacities (GSH level, TEAC level, CAT activity, and GPx activity) in HFCD fed hamsters. To sum up, those benefits could be attributed to a synergetic effect of compounds in BV.

Analysis of pecan nut (Carya illinoinensis) unsaponifiable fraction – Effect of ripening stage on phytosterols and phytostanols composition

Intidhar Bouali, Hajer Trabelsi, Wahid Herchi, Lucy Martine, et al.
Food Chemistry 164 (2014) 309–316
http://dx.doi.org/10.1016/j.foodchem.2014.05.029

Changes in 4-desmethylsterol, 4-monomethylsterol, 4,4-dimethylsterol and phytostanol composition were quantitatively and qualitatively investigated during the ripening of three varieties of Tunisian grown pecan nuts. These components have many health benefits, especially in lowering LDL-cholesterol and preventing heart disease. The phytosterol composition of whole pecan kernel was quantified by Gas Chromatography–Flame Ionization Detection (GC–FID) and identified by Gas Chromatography–Mass Spectrometry (GC–MS). Fifteen phytosterols and one phytostanol were quantified. The greatest amount of phytosterols (2852.5 mg/100 g of oil) was detected in Mahan variety at 20 weeks after the flowering date (WAFD). Moore had the highest level of phytostanols (7.3 mg/100 g of oil) at 20 WAFD. Phytosterol and phytostanol contents showed a steep decrease during pecan nut development. Results from the quantitative characterization of pecan nut oils revealed that β-sitosterol, D5-avenasterol, and campesterol were the most abundant phytosterol compounds at all ripening stages.

Association between HMW adiponectin, HMW-total adiponectin ratio and early-onset coronary artery disease in Chinese population

Ying Wang, Aihua Zheng, Yunsheng Yan, Fei Song, et al.
Atherosclerosis 235 (2014) 392-397
http://dx.doi.org/10.1016/j.atherosclerosis.2014.05.910

Objective: Adiponectin is an adipose-secreting protein that shows atheroprotective property and has inverse relation with coronary artery disease (CAD). High-molecular weight (HMW) adiponectin is reported as the active form of adiponectin. In the present study, we aimed to investigate the association between total adiponectin, HMW adiponectin, HMW-total adiponectin ratio and the severity of coronary atherosclerosis, and to compare their evaluative power for the risk of CAD. Methods: Serum levels of total and HMW adiponectin were measured in 382 early-onset CAD (EOCAD) patients and 305 matched controls undergoing coronary angiography by enzyme-linked immunosorbent assay (ELISA). Gensini score was used to evaluate the severity of coronary atherosclerosis. Results: CAD onset age was positively correlated with HMW adiponectin (r = 0.383, P < 0.001) and HMW-total adiponectin ratio (r = 0.429, P < 0.001) in EOCAD patients. Total and HMW adiponectin and HMW-total adiponectin ratio were all inversely correlated with Gensini score (r=0.417, r=0.637, r=0.578, respectively; all P < 0.001). Multivariate binary logistic regression analysis demonstrated that HMW adiponectin and HMW-total adiponectin ratio were both inversely correlated with the risk of CAD (P < 0.05). ROC analysis indicated that areas under the ROC curves of HMW adiponectin and HMW-total adiponectin ratio were larger than that of total adiponectin (P < 0.05). Conclusions: Adiponectin is cardioprotective against coronary atherosclerosis onset in EOCAD patients. HMW adiponectin and HMW-total adiponectin ratio show stronger negative associations with the severity of coronary atherosclerosis than total adiponectin does. HMW adiponectin and HMW-total adiponectin ratio are effective biomarkers for the risk of CAD in Chinese population.

Gender and age were well matched between patients and controls. EOCAD patients were tended to have a history of diabetes or hypertension, more current smoking, and more use of lipid lowering drugs. Levels of total cholesterol, LDL-c, FPG, HbA1c and triglycerides were significantly higher in the patients than in controls, while HDL-cholesterol, total adiponectin, HMW adiponectin, and HMW-total adiponectin ratio were significantly lower in the patients. EOCAD patients developed different degrees of coronary atherosclerosis, and had significantly higher levels of high-sensitivity CRP and larger circumferences of waist and hip than controls.

Spearman correlation coefficients between selected cardiovascular risk factors, Gensini score and adiponectin were significant. Total and HMW adiponectin and HMW-total adiponectin ratio were all inversely correlated with Gensini score, BMI and pack years of cigarette smoking. Total and HMW adiponectin were negatively associated with triglycerides and circumference of waist and hip. LDL-cholesterol and high-sensitivity CRP were inversely correlated with HMW adiponectin and HMW-total adiponectin ratio, while HDL-cholesterol and age were positively correlated with them. FPG was only inversely associated with HMW-total adiponectin ratio.

All participants were divided into four groups according to their Gensini score, group A (control, n = 305), group B (<20, n = 154), group C (20-40, n = 121) and group D (>40, n = 105). With the increasing of Gensini score, a stepwise downward trend was observed in levels of total and HMW adiponectin and HMW-total adiponectin ratio (P < 0.001). Specifically, total adiponectin of four groups were 1.58 (0.61-4.36) mg/ml, 1.21 (0.70-2.83) mg/ml, 1.00 (0.73-1.88) mg/ml, and 0.76 (0.37-1.19) mg/ml, respectively. Except group A with B and group B with C, the differences of pairwise comparisons among all the other groups were statistically significant (all P < 0.05). HMW adiponectin of four groups were 0.91 (0.39-3.26) mg/ml, 0.55 (0.32-1.49) mg/ml, 0.46 (0.21-0.876) mg/ml, and 0.23 (0.14-0.39) mg/ml, respectively. The differences of pairwise comparisons among all the other groups were statistically significant (all P < 0.05) except group B with C. HMW-total adiponectin ratio of four groups were 0.58 (0.31-0.81), 0.47 (0.26-0.69), 0.41 (0.24-0.57), and 0.36 (0.21-0.42), respectively. The differences of pairwise comparisons among all the other groups were statistically significant (all P < 0.05) except group B with C. In the model of multivariate binary logistic regression analysis, after adjustment for conventional cardiovascular risk factors, HMW adiponectin (OR = 0.234, P < 0.011) and HMW-total adiponectin ratio (OR = 0.138, P < 0.005) remained inversely correlated with the risk of CAD, while no significant association was observed between total adiponectin and CAD

Areas under the ROC curves were compared pairwise to identify the diagnostic power for CAD among total adiponectin, HMW adiponectin, and HMW-total adiponectin ratio. HMW adiponectin and HMW-total adiponectin ratio showed greater capability for identifying CAD than total adiponectin did (0.797 vs. 0.674, 0.806 vs. 0.674; respectively, all P < 0.05); however, no significant difference was observed between HMW and HMW-total ratio (P > 0.05).

Associations between total adiponectin, HMW adiponectin, HMW-total adiponectin ratio and the severity of coronary atherosclerosis

Associations between total adiponectin, HMW adiponectin, HMW-total adiponectin ratio and the severity of coronary atherosclerosis in EOCAD patients (evaluated by Gensini score). *P < 0.05; **P < 0.001; ***P < 0.005 by Mann-Whitney U test.

Compares diagnostic power

Compares diagnostic power

Fig. Compares diagnostic power among total adiponectin, HMW adiponectin and HMW-total adiponectin ratio for CAD by ROC curves. Diagnostic power for CAD was based on discriminating patients with or without coronary atherosclerosis. The area under the curve for HMW-total adiponectin ratio (dotted black line) was larger than that for total adiponectin (fine black line) (0.806 [95%CI 0.708-0.903] vs. 0.674 [95%CI 0.552-0.797], P < 0.05) and HMW adiponectin (bold black line) (0.806 [95%CI 0.708-0.903] vs. 0.797 [95%CI 0.706-0.888], no statistically difference). Sensitivity, specificity and optimal cut off value for them were total adiponectin (57.38%, 75.86%, 1.11 mg/ml), HMW (55.74%, 93.1%, 0.49 mg/ml) and H/T (78.69%, 75.86%, 0.52), respectively.

There are two strengths in our study. One is the precise Gensini scoring system to carefully evaluate stenosis of coronary artery or branches > 0% diameter as coronary lesion, another is the specific study subjects of EOCAD in a Chinese Han population that is particularly genetically determined and not influenced by racial/ethnic disparities. The limitations of our study lie in the interference of medications such as the effect of lipid lowering drugs on the levels of adiponectin, and cardiovascular risk factors. Smoking is a conventional cardiovascular risk factor, whose interaction with HMW adiponectin level is rarely investigated, but it has been revealed to be associated with HMW adiponectin level in men according to the study from Kawamoto R et al. We did not adjust the result for the pack/year variable in the multivariate logistic regression analysis for the limitation of small sample size of male subjects in our study. The relatively small study sample also restrained our conclusion generalizable to all populations. Future researches in larger study samples and different populations are in need to validate our findings, and to explore the association of smoking with adiponectin in male subgroup analysis, and to investigate the potential mechanisms by which adiponectin affects the progression of coronary atherosclerosis.

In summary, the present study has demonstrated that adiponectin is protective against coronary atherosclerosis onset in EOCAD patients. HMW adiponectin and HMW-total adiponectin ratio show stronger negative associations with the severity of coronary atherosclerosis than total adiponectin does. HMW adiponectin and HMW-total adiponectin ratio are more effective biomarkers for the risk of CAD than total adiponectin.

Berberis aristata combined with Silybum marianum on lipid profile in patients not tolerating statins at high doses

Giuseppe Derosa, Davide Romano, Angela D’Angelo, Pamela Maffioli
Atherosclerosis 239 (2015) 87-92
http://dx.doi.org/10.1016/j.atherosclerosis.2014.12.043

Aim: To evaluate the effects of Berberis aristata combined with Silybum marianum in dyslipidemic patients intolerant to statins at high doses.
Methods: 137 euglycemic, dyslipidemic subjects, with previous adverse events to statins at high doses, were enrolled. Statins were stopped for 1 month (run-in), then they were re-introduced at the half of the previously taken dose. At randomization, patients tolerating the half dose of statin, were assigned to
add placebo or B. aristata/S. marianum 588/105 mg, 1 tablet during the lunch and 1 tablet during the dinner, for six months. We evaluated lipid profile and safety parameters variation at randomization, and after 3, and 6 months.
Results: B. aristata/S. marianum reduced fasting plasma glucose (-9 mg/dl), insulin (-0.7 mU/ml), and HOMA-index (-0.35) levels compared to baseline and also to placebo. Lipid profile did not significantly change after 6 months since the reduction of statin dosage and the introduction of B. aristata/S. marianum, while it worsened in the placebo group both compared to placebo and with active treatment (+23.4 mg/dl for total cholesterol, +19.6 mg/dl for LDL-cholesterol, +23.1 mg/dl for triglycerides with placebo compared to B. aristata/S. marianum). We did not record any variations of safety parameters
in either group. Conclusions: B. aristata/S. marianum can be considered as addition to statins in patients not tolerating high dose of these drugs.

Statins, also known as 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, are effective medications for reducing the risk of death and future cardiovascular disease. In the latest years, however, statin intolerance (including adverse effects related to quality of life, leading to decisions to decrease or stop the use of an otherwise-beneficial drug) has come to the forefront of clinical concern, whereas the safety of statins has come to be regarded as largely favorable. Statin intolerance is defined as any adverse symptoms, signs, or laboratory abnormalities attributed by the patient or physician to the statin and in most cases perceived by the patient to interfere unacceptably with activities of daily living, leading to a decision to stop or reduce statin therapy. The physician might also decide to stop or reduce statin therapy on the basis of clinical/laboratory assessment [abnormal liver function tests, creatine phosphokinase values (CPK)] suggesting undue risk. Adverse events are more common at higher doses of statins, and often contribute to patients low adherence to treatment. For this reason, researchers are testing alternative strategies for lipid treatment when statin intolerance is recognized. One strategy to reduce the risk of statin-induced adverse events includes using a low-dose of statin combined with nonstatin drugs in order to achieve the goals of therapy. Nonstatin drugs include nutraceuticals; in the latest years relatively large number of dietary supplements and nutraceuticals have been studied for their supposed or demonstrated ability to reduce cholesterolemia in humans, in particular Berberis Aristata, has been studied in randomized clinical trials and proved to be effective in improving lipid profile. In particular, B. aristata acts up-regulating LDL-receptor (LDL-R) expression independent of sterol regulatory element binding proteins, but dependent on extracellular signal-regulated kinases (ERK) and c-Jun N-terminal kinase (JNK) activation leading to total cholesterol (TC) and LDL-C reduction of about 30 and 25%, respectively. Hwever, B. aristata is a problem in terms of oral bioavailability, affected by a P-glycoprotein (P-gp) mediated gut extrusion process. P-gp seems to reduce by about 90% the amount of B. aristata able to cross the enterocytes, but the use of a potential P-gp inhibitor could ameliorate its oral poor bioavailability improving its effectiveness. Among the potential Pgp inhibitors, silymarin from S. marianum, an herbal drug used as liver protectant, could be considered a good candidate due to its high safety profile.

Analyzing the results of our study, it can appear, at a first glance, that B. aristata/S. marianum has a neutral effect of lipid profile that did not change during the study after the addition of the nutraceutical combination. This lack of effect, however, is only apparent, because, when we analyzed what happens in placebo group, we observed a worsening of lipid profile after statin dose reduction. In other words, the addition of B. aristata/S. marianum neutralized the worsening of lipid profile observed with placebo after statins dose reduction. These results are in line with what was reported by Kong et al., who evaluated the effects of a combination of berberine and simvastatin in sixty-three outpatients diagnosed with hypercholesterolemia. As compared with monotherapies, the combination showed an improved lipid lowering effect with 31.8% reduction of serum LDL-C, and similar efficacies were observed in the reduction of TC as well as Tg in patients. Considering the results of this study, B. aristata/S. marianum can be considered as addition to statins in patients not tolerating high dose of these drugs.

CETP inhibitors downregulate hepatic LDL receptor and PCSK9 expression in vitro and in vivo through a SREBP2 dependent mechanism

Bin Dong, Amar Bahadur Singh, Chin Fung, Kelvin Kan, Jingwen Liu
Atherosclerosis 235 (2014) 449-462
http://dx.doi.org/10.1016/j.atherosclerosis.2014.05.931

Background: CETP inhibitors block the transfer of cholesteryl ester from HDL-C to VLDL-C and LDL-C, thereby raising HDL-C and lowering LDL-C. In this study, we explored the effect of CETP inhibitors on hepatic LDL receptor (LDLR) and PCSK9 expression and further elucidated the underlying regulatory mechanism. Results: We first examined the effect of anacetrapib (ANA) and dalcetrapib (DAL) on LDLR and PCSK9 expression in hepatic cells in vitro. ANA exhibited a dose-dependent inhibition on both LDLR and PCSK9 expression in CETP-positive HepG2 cells and human primary hepatocytes as well as CETP-negative mouse primary hepatocytes (MPH). Moreover, the induction of LDLR protein expression by rosuvastatin in MPH was blunted by cotreatment with ANA. In both HepG2 and MPH ANA treatment reduced the amount of mature form of SREBP2 (SREBP2-M). In vivo, oral administration of ANA to dyslipidemic C57BL/6J mice at a daily dose of 50 mg/kg for 1 week elevated serum total cholesterol by approximately 24.5% (p < 0.05%) and VLDL-C by 70% (p < 0.05%) with concomitant reductions of serum PCSK9 and liver LDLR/SREBP2-M protein. Finally, we examined the in vitro effect of two other strong CETP inhibitors evacetrapib and torcetrapib on LDLR/PCSK9 expression and observed a similar inhibitory effect as ANA in a concentration range of 1-10 µM. Conclusion: Our study revealed an unexpected off-target effect of CETP inhibitors that reduce the mature form of SREBP2, leading to attenuated transcription of hepatic LDLR and PCSK9. This negative regulation of SREBP pathway by ANA manifested in mice where CETP activity was absent and affected serum cholesterol metabolism.

Effect of Eclipta prostrata on lipid metabolism in hyperlipidemic animals

Yun Zhao, Lu Peng, Wei Lu, Yiqing Wang, Xuefeng Huang, et al.
Experimental Gerontology 62 (2015) 37–44
http://dx.doi.org/10.1016/j.exger.2014.12.017

Eclipta prostrata (Linn.) Linn. is a traditional Chinese medicine and has previously been reported to have hypolipidemic effects. However, its mechanism of action is not well understood. This study was conducted to identify the active fraction of Eclipta, its toxicity, its effect on hyperlipidemia, and its mechanism of action. The ethanol extract (EP) of Eclipta and fractions EPF1–EPF4, obtained by eluting with different concentrations of ethanol from a HPD-450 macroporous resin column chromatography of the EP, were screened in hyperlipidemic mice for lipid lowering activity, and EPF3 was the most active fraction. The LD50 of EPF3 was undetectable because no mice died with administration of EPF3 at 10.4 g/kg. Then, 48 male hamsters were used and randomly assigned to normal chow diet, high-fat diet, high-fat diet with Xuezhikang (positive control) or EPF3 (75, 150 and 250 mg/kg) groups. We evaluated the effects of EPF3 on body weight gain, liver weight gain, serum lipid concentration, antioxidant enzyme activity, and the expression of genes involved in lipid metabolism in hyperlipidemic hamsters. The results showed that EPF3 significantly decreased body-weight gain and liver-weight gain and reduced the serum lipid levels in hyperlipidemic hamsters. EPF3 also increased the activities of antioxidant enzymes; upregulated the mRNA expression of peroxisome proliferator-activated receptor α (PPARα), low density lipoprotein receptor (LDLR), lecithin-cholesterol transferase (LCAT) and scavenger receptor class B type Ι receptor (SR-BI); and down-regulated the mRNA expression of 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGR) in the liver. These results indicate that EPF3 ameliorates hyperlipidemia, in part, by reducing oxidative stress and modulating the transcription of genes involved in lipid metabolism.

Although Eclipta has long been used as a food additive, no studies or reports have clearly shown any liver or kidney toxicity from its use. Therefore, E. prostrata is safe and beneficial for preventing hyperlipidemia in experimental animals and can be used as an alternative medicine for the regulation of dyslipidemia.

Effect of high fiber products on blood lipids and lipoproteins in hamsters

HE Martinez-Floresa, Y Kil Chang, F Martinez-Bustosc, V Sgarbieri
Nutrition Research 24 (2004) 85–93
http://dx.doi.org:/10.1016/S0271-5317(03)00206-9

Serum and liver lipidemic responses in hamsters fed diets containing 2% cholesterol and different dietary fiber sources were studied. The following diets were made from: a) the control diet made from extruded cassava starch (CSH) contained 9.3% cellulose, b) cassava starch extruded with 9.7% resistant starch (CS-RS), c) cassava starch extruded with 9.9% oat fiber (CS-OF), d) the reference diet contained 9.5% cellulose, and no cholesterol was added. Total cholesterol, LDLVLDL-cholesterol and triglycerides were significantly lower (P < 0.05) in serum of hamsters fed on the CS-RS (17.87%, 62.92% and 9.17%, respectively) and CS-OF (15.12%, 67.41% and 18.35%, respectively) diets, as compared to hamster fed with the CSH diet. Similar results were found in the livers of hamsters fed on the CS-RS and CS-OF diets, as compared to hamsters fed with the CSH diet. The diets containing these fibers could be used as active ingredients in human diets to improve the human health.

A new piece in the puzzling effect of n-3 fatty acids on atherosclerosis?

Wilfried Le Goff
Atherosclerosis 235 (2014) 358-362
http://dx.doi.org/10.1016/j.atherosclerosis.2014.03.038

Omega-3 fatty acids (ω-3) FA are reported to be protective against cardiovascular disease (CVD), notably through their beneficial action on atherosclerosis development. In this context dietary intake of long chain marine eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) is recommended and randomised trials largely support that EPA and DHA intake is associated with a reduction of CVD. However, mechanisms governing the atheroprotective action of ω-3 FA are still unclear and numerous studies using mouse models conducted so far do not allow to reach a precise view of the cellular and molecular effects of ω-3 FA on atherosclerosis. In the current issue of Atherosclerosis, Chang et al. provide important new information on the anti-atherogenic properties of ω-3 FA by analyzing the incremental replacement of saturated FA by pure fish oil as a source of EPA and DHA in Ldlr -/- mice fed a high fat/high cholesterol diet.

Cardiovascular disease (CVD) is the leading causes of death in the world and is frequently associated with atherosclerosis, a pathology characterized by the accumulation of lipids, mainly cholesterol in the arterial wall. Among major risk factors for CVD, circulating levels of lipids and more especially those originating from diets are closely linked to development of atherosclerosis. In this context, not only cholesterol, but also dietary fatty acids (FA) may appear particularly deleterious in regards to atherosclerosis and associated CVD. However, although saturated fats are proatherogenic, omega-3 fatty acids (ω-3 FA), and more especially long-chain marine eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), exert atheroprotective properties through several potential underlying mechanisms. Therefore, the intake of EPA and DHA is recommended around the world and randomised trials with ω-3 FA confirmed that EPA and DHA intake reduced risk for CVD events. However benefits of ω-3 FA intake were challenged by recent clinical trials that failed to replicate protective effects of EPA + DHA on CVD, raising the controversy on the healthy side of marine ω-3 FA.

Animal models are commonly employed in order to decipher mechanisms by which ω-3 FA exert their beneficial actions regarding lipid metabolism and atherosclerosis. Since the last past 20 years, mouse models, and more especially genetically modified mouse models, became the reference model to evaluate the effects of dietary fatty acids, especially ω-3 FA, on atherosclerosis development [7-20]. However, the use of different mouse models of atherosclerosis (Apoe-/-, Ldlr-/-, double Apoe-/- x Ldlr-/- , Ldlr-/- x hApoB mice), as well as diet composition (chow, high cholesterol, high fat, high cholesterol/high fat), source of ω-3 FA supplementation (fish oil, perilla seed oil, flaxseed, pure ALA, EPA or DHA), duration of the diet (from 4 to 32 weeks), size of atherosclerotic lesions in control animals (from 51 to 700.103 mm2) in

those studies led to heterogeneous results and therefore to a partial understanding of the effects of ω-3 FA on atherosclerosis.

Contrary to what observed in Apoe-/- mice, dietary supplementation of Ldlr-/- mice with ω-3 FA led to a reproducible reduction of aortic atherosclerosis, although to various degrees, confirming that Ldlr-/- mice constitute the most appropriate model for studying the atheroprotective effects of ω-3 FA. When evaluated, the decrease of atherosclerosis upon ω-3 FA-rich diet was accompanied by a reduction in the macrophage content as well as inflammation in aortic lesions highlighting the major impact of ω-3 FA on monocyte recruitment and subsequent macrophage accumulation in the arterial wall. However, although supplementation with ω-3 FA allows an efficacious lowering of plasma lipid levels in humans, studies in mouse models suggest that the antiatherogenic action of ω-3 FA is independent of any effects on plasma cholesterol or triglyceride levels. However, that must be asserted with caution as lipid metabolism is quite different in mouse in comparison to humans, highlighting the need to study in the future the effects of ω-3 FA on atherosclerosis in a mouse model exhibiting a more “humanized” lipid metabolism as achieved in hApoB/CETP mice.

In a previous issue of Atherosclerosis, Chang et al. reevaluate the impact of fish oil ω-3 FA on atherosclerosis development by operating an incremental replacement of saturated fats (SAT) by ω-3 FA (pure fish oil, EPA- and DHA-rich) in Ldlr-/- mice fed a high-fat (21%, w/w)/high-cholesterol (0.2%, w/w) diet for a 12-week period. This experimental approach is quite pertinent as dietary fat intake in developed countries, as in United States, derived mostly from saturated FA and is poor in ω-3 FA. Then, using this strategy the authors were able to evaluate the potential beneficial effects of a supplementation with fish oil ω-3 FA in a dietary context for which ω-3 FA intake is relevant.

Here, Chang et al. demonstrated that the progressive increase of dietary intake of fish oil ω-3 FA (EPA and DHA) abrogated the deleterious effects of a SAT diet, thereby suggesting that a dietary ω-3 FA intake on a SAT background is potentially efficient to decrease CVD in humans. Indeed, replacement of SAT by fish oil ω-3 FA markedly reduced plasma cholesterol and triglycerides levels and abolished diet-induced atherosclerosis mediated by SAT in Ldlr-/-mice. To note that in the present study, Ldlr-/- mice only developed small atherosclerosic lesions (~100.103 mm2) after 12 weeks of diet with SAT.

As previously reported, decreased atherosclerotic lesions were accompanied by a reduced content of aortic macrophages and inflammation. Based on their previous works, the authors proposed that the reduction of atherosclerosis upon ω-3 FA resulted from an impairment of cholesterol uptake by arterial macrophages consecutive to the decrease of Lipoprotein Lipase (LPL) expression in those cells. Indeed, beyond its lipolysis action on triglycerides, LPL was reported to promote lipid accumulation, in particular in macrophages, by binding to lipoproteins and cell surface proteoglycans and then acting as a bridging molecule that facilitates cellular lipid uptake. Coherent with this mechanism, macrophage LPL expression was reported to promote foam cell formation and atherosclerosis. In the present study, replacement of SAT by ω-3 FA both decreased expression and altered distribution of arterial LPL. Such a mechanism for ω-3 FA (EPA and DHA) was proposed by this group in earlier studies to favor reduction of arterial LDL-cholesterol. It is noteworthy that lipid rafts alter distribution of LPL at the cell surface and subsequently the LPL dependent accumulation of lipids in macrophages and foam cell formation. As incorporation of ω-3 FA, such as DHA, into cell membrane phospholipids disrupts lipid rafts organization, it cannot be exclude that reduction of lipid accumulation in arterial macrophages upon addition of ω-3 FA results in part from an impairment of the localization and of the anchoring function of LPL at the cell surface of macrophages. Indeed Chang et al. observed that progressive replacement of SAT by ω-3 FA affected aortic FA composition leading to a pronounced increase of arterial EPA and DHA, then suggesting that content of ω-3 FA in macrophage membrane may be equally altered. However, the implication of LPL in the atheroprotective effects of ω-3 FA need to be validated using an appropriate mouse model for which LPL expression may be controlled.

Among the various mechanisms by which ω-3 FA exert anti-inflammatory properties, EPA and DHA repressed inflammation by shutting down NF-kB activation in macrophages. Since expression of TLR-4 and NF-kB target genes, IL-6 and TNFα, in aorta from mice fed diets containing ω-3 FA were decreased when compared to SAT, those results strongly support the contention that ω-3 FA repress inflammation by inhibiting the TLR4/NF-kB signaling cascade likely through the macrophage ω-3 FA receptor GPR120.

Although further studies are needed to explore the complete spectrum of actions of ω-3 FA on atherosclerosis development and CVD, this study provides important information that supports that ω-3 FA intake is a pertinent strategy to reduce risk of CVD.

Effects of dietary hull-less barley β-glucan on the cholesterol metabolism of hypercholesterolemic hamsters

Li-Tao Tong, Kui Zhong, Liya Liu, Xianrong Zhou, Ju Qiu, Sumei Zhou
Food Chemistry 169 (2015) 344–349
http://dx.doi.org/10.1016/j.foodchem.2014.07.157

The aim of the present study is to investigate the hypocholesterolemic effects of dietary hull-less barley β-glucan (HBG) on cholesterol metabolism in hamsters which were fed a hypercholesterolemic diet. The hamsters were divided into 3 groups and fed experimental diets, containing 5‰ HBG or 5‰ oat β-glucan (OG), for 30 days. The HBG, as well as OG, lowered the concentration of plasma LDL-cholesterol significantly. The excretion of total lipids and cholesterol in feces were increased in HBG and OG groups compared with the control group. The activity of 3-hydroxy-3-methyl glutaryl-coenzyme A (HMG-CoA) reductase in liver was reduced significantly in the HBG group compared with the control and OG groups. The activity of cholesterol 7-α hydroxylase (CYP7A1) in the liver, in the HBG and OG groups, was significantly increased compared with the control group. The concentrations of acetate, propionate and total short chain fatty acids (SCFAs) were not significantly different between the HBG and control groups. These results indicate that dietary HBG reduces the concentration of plasma LDL cholesterol by promoting the excretion of fecal lipids, and regulating the activities of HMG-CoA reductase and CYP7A1 in hypercholesterolemic hamsters.

Effects of dietary wheat bran arabinoxylans on cholesterolmetabolism of hypercholesterolemic hamsters

Li-Tao Tong, Kui Zhong, Liya Liu, Ju Qiu, Lina Guo, et al.
Carbohydrate Polymers 112 (2014) 1–5
http://dx.doi.org/10.1016/j.carbpol.2014.05.061

The aim of the present study is to investigate the effects of dietary wheat bran arabinoxylans (AXs) on cholesterol metabolism in hypercholesterolemic hamsters. The hamsters were divided into 3 groups and fed the experimental diets containing AXs or oat β-glucan at a dose of 5 g/kg for 30 days. As the results,the AXs lowered plasma total cholesterol and LDL-cholesterol concentrations, and increased excretions of total lipids, cholesterol and bile acids, as well as oat β-glucan. The AXs reduced the activity of 3-hydroxy-3-methyl glutaryl-coenzyme A (HMG-CoA) reductase, and increased the activity of cholesterol 7-α hydroxylase (CYP7A1) in liver. Moreover, the AXs increased propionate and the total short-chain fatty acids (SCFAs) concentrations. These results indicated that dietary AXs reduced the plasma total cholesterol and LDL-cholesterol concentrations by promoting the excretion of fecal lipids, regulating the activities of HMG-CoA reductase and CYP7A1, and increasing colonic SCFAs in hamsters.

High-fructose feeding promotes accelerated degradation of hepatic LDL receptor and hypercholesterolemia in hamsters via elevated circulating PCSK9 levels

Bin Dong, Amar Bahadur Singh, Salman Azhar, Nabil G. Seidah, Jingwen Liu
Atherosclerosis 239 (2015) 364-374
http://dx.doi.org/10.1016/j.atherosclerosis.2015.01.013

Background: High fructose diet (HFD) induces dyslipidemia and insulin resistance in experimental animals and humans with incomplete mechanistic understanding. By utilizing mice and hamsters as in vivo models, we investigated whether high fructose consumption affects serum PCSK9 and liver LDL receptor (LDLR) protein levels. Results: Feeding mice with an HFD increased serum cholesterol and reduced serum PCSK9 levels as compared with the mice fed a normal chow diet (NCD). In contrast to the inverse relationship in mice, serum PCSK9 and cholesterol levels were co-elevated in HFD-fed hamsters. Liver tissue analysis revealed that PCSK9 mRNA and protein levels were both reduced in mice and hamsters by HFD feeding, however, liver LDLR protein levels were markedly reduced by HFD in hamsters but not in mice. We further showed that circulating PCSK9 clearance rates were significantly lower in hamsters fed an HFD as compared with the hamsters fed NCD, providing additional evidence for the reduced hepatic LDLR function by HFD consumption. The majority of PCSK9 in hamster serum was detected as a 53 kDa N-terminus cleaved protein. By conducting in vitro studies, we demonstrate that this 53 kDa truncated hamster PCSK9 is functionally active in promoting hepatic LDLR degradation. Conclusion: Our studies for the first time demonstrate that high fructose consumption increases serum PCSK9 concentrations and reduces liver LDLR protein levels in hyper-lipidemic hamsters. The positive correlation between circulating cholesterol and PCSK9 and the reduction of liver LDLR protein in HFD-fed hamsters suggest that hamster is a better animal model than mouse to study the modulation of PCSK9/LDLR pathway by atherogenic diets.

High-oleic canola oil consumption enriches LDL particle cholesteryl oleate content and reduces LDL proteoglycan binding in humans

Peter J.H. Jones, Dylan S. MacKay, Vijitha K. Senanayake, Shuaihua Pu, et al.
Atherosclerosis 238 (2015) 231-238
http://dx.doi.org/10.1016/j.atherosclerosis.2014.12.010

Oleic acid consumption is considered cardio-protective according to studies conducted examining effects of the Mediterranean diet. However, animal models have shown that oleic acid consumption increases LDL particle cholesteryl oleate content which is associated with increased LDL-proteoglycan binding and atherosclerosis. The objective was to examine effects of varying oleic, linoleic and docosahexaenoic acid consumption on human LDL-proteoglycan binding in a non-random subset of the Canola Oil Multi-center Intervention Trial (COMIT) participants. COMIT employed a randomized, double-blind, five-period, crossover trial design. Three of the treatment oil diets: 1) a blend of corn/safflower oil (25:75); 2) high oleic canola oil; and 3) DHA-enriched high oleic canola oil were selected for analysis of LDL-proteoglycan binding in 50 participants exhibiting good compliance. LDL particles were isolated from frozen plasma by gel filtration chromatography and LDL cholesteryl esters quantified by mass-spectrometry. LDL-proteoglycan binding was assessed using surface plasmon resonance. LDL particle cholesterol ester fatty acid composition was sensitive to the treatment fatty acid compositions, with the main fatty acids in the treatments increasing in the LDL cholesterol esters. The corn/safflower oil and high-oleic canola oil diets lowered LDL-proteoglycan binding relative to their baseline values (p < 0.0005 and p < 0.0012, respectively). At endpoint, high-oleic canola oil feeding resulted in lower LDL-proteoglycan binding than corn/safflower oil (p < 0.0243) and DHA-enriched high oleic canola oil (p < 0.0249), although high-oleic canola oil had the lowest binding at baseline (p < 0.0344). Our findings suggest that high-oleic canola oil consumption in humans increases cholesteryl oleate percentage in LDL, but in a manner not associated with a rise in LDL-proteoglycan binding.

Read Full Post »

Acute Lung Injury

Writer and Curator: Larry H. Bernstein, MD, FCAP 

 

 

Introduction

Acute lung injury is a serious phenomenon only recognized as having significant relevance to allogeneic blood transfusion in the last 15 years.  It is not limited to transfusion events, and is also related to SIRS and sepsis.  It is simulated in experimental models by lipoprotein, such as endotoxin.  It occurs in the pretransfused surgical patient, or in the medical patient as well.  Why it was not recognized earlier is a matter of conjecture.  The significant reduction in immune modulated blood type incompatibility reactions in Western countries is a factor.  The other factor is that the lipoprotein antigenic fractions involved are associated with component transfusions other than stored red cells. The following discussion will elaborate on what is increasingly recognized as a relevant issue in medicine today.
Transfusion Related Reaction

In medicinetransfusion related acute lung injury (TRALI) is a serious blood transfusion complication characterized by the acute onset of non-cardiogenic pulmonary edema following transfusion of blood products.[1]

Although the incidence of TRALI has decreased with modified transfusion practices, it is still the leading cause of transfusion-related fatalities in the United States from fiscal year 2008 through fiscal year 2012.

Transfusion Related Acute Lung Injury

TRALI-Hyaline_membranes_-_very_high_mag

TRALI-Hyaline_membranes_-_very_high_mag

Micrograph of diffuse alveolar damage, the histologic correlate of TRALI. H&E stain. Very high magnification micrograph of hyaline membranes, as seen in diffuse alveolar damage (DAD), the histologic correlate of acute respiratory distress syndrome (ARDS), transfusion related acute lung injury (TRALI), acute interstitial pneumonia (AIP).
http://upload.wikimedia.org/wikipedia/commons/thumb/c/c8/Hyaline_membranes_-_very_high_mag.jpg/1024px-Hyaline_membranes_-_very_high_mag.jpg

TRALI is defined as an acute lung injury that is temporally related to a blood transfusion; specifically, it occurs within the first six hours following a transfusion.[3]

It is typically associated with plasma components such as platelets and Fresh Frozen Plasma, though cases have been reported with packed red blood cells since there is some residual plasma in the packed cells. The blood component transfused is not part of the case definition. Transfusion-related acute lung injury (TRALI) is an uncommon syndrome that is due to the presence of leukocyte antibodies in transfused plasma. TRALI is believed to occur in approximately one in every 5000 transfusions. Leukoagglutination and pooling of granulocytes in the recipient’s lungs may occur, with release of the contents of leukocyte granules, and resulting injury to cellular membranes, endothelial surfaces, and potentially to lung parenchyma. In most cases leukoagglutination results in mild dyspnea and pulmonary infiltrates within about 6 hours of transfusion, and spontaneously resolves;

Occasionally more severe lung injury occurs as a result of this phenomenon and Acute Respiratory Distress Syndrome (ARDS) results. Leukocyte filters may prevent TRALI for those patients whose lung injury is due to leukoagglutination of the donor white blood cells, but because most TRALI is due to donor antibodies to leukocytes, filters are not helpful in TRALI prevention. Transfused plasma (from any component source) may also contain antibodies that cross-react with platelets in the recipient, producing usually mild forms of posttransfusion purpura or platelet aggregation after transfusion.

Another nonspecific form of immunologic transfusion complication is mild to moderate immunosuppression consequent to transfusion. This effect of transfusion is not completely understood, but appears to be more common with cellular transfusion and may result in both desirable and undesirable effects. Mild immunosuppression may benefit organ transplant recipients and patients with autoimmune diseases; however, neonates and other already immunosuppressed hosts may be more vulnerable to infection, and cancer patients may possibly have worse outcomes postoperatively.

http://en.wikipedia.org/wiki/Transfusion-related_acute_lung_injury

 

 

Perioperative transfusion-related acute lung injury: The Canadian Blood Services experience

Asim Alam, Mary Huang, Qi-Long Yi, Yulia Lin, Barbara Hannach
Transfusion and Apheresis Science 50 (2014) 392–398
http://dx.doi.org/10.1016/j.transci.2014.04.008

Purpose: Transfusion-related acute lung injury (TRALI) is a devastating transfusion-associated adverse event. There is a paucity of data on the incidence and characteristics of TRALI cases that occur perioperatively. We classified suspected perioperative TRALI cases reported to Canadian Blood Services between 2001 and 2012, and compared them to non-perioperative cases to elucidate factors that may be associated with an increased risk of developing TRALI in the perioperative setting. Methods: All suspected TRALI cases reported to Canadian Blood Services (CBS) since 2001 were reviewed by two experts or, from 2006 to 2012, the CBS TRALI Medical Review Group (TMRG). These cases were classified based on the Canadian Consensus Conference (CCC) definitions and detailed in a database. Two additional reviewers further categorized them as occurring within 72 h from the onset of surgery (perioperative) or not in that period (non-perioperative). Various demographic and characteristic variables of each case were collected and compared between groups. Results: Between 2001 and 2012, a total of 469 suspected TRALI cases were reported to Canadian Blood Services; 303 were determined to be within the TRALI diagnosis spectrum. Of those, 112 (38%) were identified as occurring during the perioperative period. Patients who underwent cardiac surgery requiring cardiopulmonary bypass (25.0%), general surgery (18.0%) and orthopedics patients (12.5%) represented the three largest surgical groups. Perioperative TRALI cases comprised more men (53.6% vs. 41.4%, p = 0.04) than non-perioperative patients. Perioperative TRALI patients more often required supplemental O2 (14.3% vs. 3.1%, p = 0.0003), mechanical ventilation (18.8% vs. 3.1%), or were in the ICU (14.3% vs. 3.7%, p = 0.0043) prior to the onset of TRALI compared to non-perioperative TRALI patients. The surgical patients were transfused on average more components than non-perioperative patients (6.0 [SD = 8.3] vs. 3.6 [5.2] products per patient, p = 0.0002). Perioperative TRALI patients were transfused more plasma (152 vs. 105, p = 0.013) and cryoprecipitate (51 vs. 23, p < 0.01) than non-perioperative TRALI patients. There was no difference between donor antibody test results between the groups. Conclusion: CBS data has provided insight into the nature of TRALI cases that occur perioperatively; this  group represents a large proportion of TRALI cases.

 

Transfusion-related acute lung injury: a clinical review

Alexander P J Vlaar, Nicole P Juffermans
Lancet 2013; 382: 984–94
http://dx.doi.org/10.1016/S0140-6736(12)62197-7

Three decades ago, transfusion-related acute lung injury (TRALI) was considered a rare complication of transfusion medicine. Nowadays, the US Food and Drug Administration acknowledge the syndrome as the leading cause of transfusion-related mortality. Understanding of the pathogenesis of TRALI has resulted in the design of preventive strategies from a blood-bank perspective. A major breakthrough in efforts to reduce the incidence of TRALI has been to exclude female donors of products with high plasma volume, resulting in a decrease of roughly two-thirds in incidence. However, this strategy has not completely eradicated the complication. In the past few years, research has identified patient-related risk factors for the onset of TRALI, which have empowered physicians to take an individualized approach to patients who need transfusion.

Development of an international consensus definition has aided TRALI research, yielding a higher incidence in specific patient populations than previously acknowledged Patients suffering from a clinical disorder such as sepsis are increasingly recognized as being at risk for development of TRALI. Thereby, from a diagnosis by exclusion, TRALI has become the leading cause of transfusion-related mortality. However, the syndrome is still under diagnosed and under-reported in some countries.

Although blood transfusion can be life-saving, it can also be a life-threatening intervention. Physicians use blood transfusion on a daily basis. Increased awareness of the risks of this procedure is needed, because management of patient-tailored transfusion could reduce the risk of TRALI. Such an individualized approach is now possible as insight into TRALI risk factors evolves. Furthermore, proper reporting of TRALI could prevent recurrence.

Absence of an international definition for TRALI previously contributed to underdiagnosis. As such, a consensus panel, and the US National Heart, Lung and Blood Institute Working Group in 2004, formulated a case definition of TRALI based on clinical and radiological parameters. The definition is derived from the widely used definition of acute lung injury (panel 1). Suspected TRALI is defined as fulfilment of the definition of acute lung injury within 6 h of transfusion in the absence of another risk factor (panel 1).

Although this definition seems to be straightforward, the characteristics of TRALI are indistinguishable from acute lung injury due to other causes, such as sepsis or lung contusion. Therefore, this definition would rule out the possibility of diagnosing TRALI in a patient with an underlying risk factor for acute lung injury who has also received a transfusion. To identify such cases, the term possible TRALI was developed.

Although the TRALI definition is an international consensus definition, surveillance systems in some countries, including the USA, France and the Netherlands, use an alternative in which imputability is scored. Imputability aims to identify the likelihood that transfusion is the causal factor. Imputability scores mostly imply that other causes of acute lung injury can be ruled out, so that diagnosis of TRALI is by exclusion. However, observational and animal studies suggest that risk factors for TRALI include other disorders, such as sepsis. Therefore, an imputability definition would result in underdiagnosis of TRALI. The consensus definition accommodates the uncertainty of the association of acute lung injury to the transfusion in possible TRALI. The conventional definition of TRALI uses a timeframe of 6 h in which acute lung injury needs to develop after a blood transfusion. In critically ill patients, transfusion increases the risk (odds ratio 2·13, 95% CI 1·75–2·52) for development of acute lung injury 6–72 h after transfusion.  However, whether the pathogenesis of delayed TRALI is similar to that of TRALI is unclear.

A two-hit hypothesis has been proposed for TRALI. The first hit is underlying patient factors, resulting in adherence of primed neutrophils to the pulmonary endothelium. The second hit is caused by mediators in the blood transfusion that activate the endothelial cells and pulmonary neutrophils, resulting in capillary leakage and subsequent pulmonary edema. The second hit can be antibody-mediated or non-antibody-mediated.

Panel 1: Definition of transfusion-related acute lung injury (TRALI)

Suspected TRALI

  • Acute onset within 6 h of blood transfusion
    • PaO2/FIO2<300 mm Hg, or worsening of P to F ratio
    • Bilateral infi ltrative changes on chest radiograph
    • No sign of hydrostatic pulmonary oedema (pulmonary arterial occlusion
    pressure ≤18 mm Hg or central venous pressure ≤15 mm Hg)
    • No other risk factor for acute lung injury

Possible TRALI
Same as for suspected TRALI, but another risk factor present for acute lung injury

Delayed TRALI
Same as for (possible) TRALI and onset within 6–72 h of blood transfusion

Pathophysiology of two-hit mediated transfusion-related acute lung injury (TRALI).  The pre-phase of the syndrome consists of a fi rst hit, which is mainly systemic. This first hit is the underlying disorder of the patient (eg, sepsis or pneumonia) causing neutrophil attraction to the capillary of the lung. Neutrophils are attracted to the lung by release of cytokines and chemokines from upregulated lung endothelium. Loose binding by L-selectin takes place. Firm adhesion is mediated by E-selectin and platelet-derived P-selectin and intracellular adhesion molecules (ICAM-1). In the acute phase of the syndrome, a second hit caused by mediators in the blood transfusion takes place. This hit results in activation of inflammation and coagulation in the pulmonary compartment. Neutrophils adhere to the injured capillary endothelium and marginate through the interstitium into the air space, which is filled with protein-rich edema fluid. In the air space, cytokines interleukin-1, -6, and -8, (IL-1, IL-6, and IL-8, respectively) are secreted, which act locally to stimulate chemotaxis and activate neutrophils resulting in formation of the elastase-α1-antitrypsin (EA) complex. Neutrophils can release oxidants, proteases, and other proinflammatory molecules, such as platelet-activating factor (PAF), and form neutrophil extracellular traps (NETs). Furthermore, activation of the coagulation system happens, shown by an increase in thrombin-antithrombin complexes (TATc), as does a decrease in activity of the fibrinolysis system, shown by a reduction in plasminogen activator activity. The influx of protein-rich edema fluid into the alveolus leads to the inactivation of surfactant, which contributes to the clinical picture of acute respiratory distress in the onset of TRALI. PAI-1 = plasminogen activator inhibitor-1.

Antibody-mediated TRALI is caused by passive transfusion of HLA or human neutrophil antigen (HNA) and corresponding antibodies from the donor directed against antigens of the recipient. Neutrophil activation occurs directly by binding of the antibody to the neutrophil surface (HNA antibodies) or indirectly, mainly by binding to the endothelial cells with activation of the neutrophil (HLA class I antibodies) or to monocytes with subsequent activation of the neutrophil (HLA class II antibodies). The antibody titer and the volume of antibody containing plasma both increase the risk for onset of TRALI. Although the role of donor HLA and HNA antibodies from transfused blood is widely accepted, not all TRALI cases are antibody mediated. In many patients, antibodies cannot be detected. Furthermore, many blood products containing antibodies do not lead to TRALI. This finding has led to development of an alternative hypothesis for the onset of TRALI, termed non-antibody-mediated TRALI.

Non-antibody-mediated TRALI is caused by accumulation of proinflammatory mediators during storage of blood products, and possibly by ageing of the erythrocytes and platelets themselves. Although most preclinical studies have noted a positive correlation between storage time of cell-containing blood products and TRALI, the mechanism is controversial. Two mechanisms have been suggested, including either plasma or the aged cells. In a small-case study and animal experiments, accumulation of bioactive lipids and soluble CD40 ligand (sCD40L) in the plasma layer of cell-containing blood products has been associated with TRALI. Bioactive lipids are thought to cause neutrophil activation through the G-protein coupled receptor on the neutrophil.

The two-hit model suggests that patients in a poor clinical state are at risk for development of TRALI. However, cases have been described of antibody-mediated TRALI developing in fairly healthy recipients. To explain this discrepancy, a threshold model has been suggested in which a threshold must be overcome to induce a TRALI reaction. The threshold is dependent both on the predisposition of the patient (first hit) and the quantity of antibodies in the transfusion (second hit). A large quantity of antibody that matches the recipient’s antigen can cause severe TRALI in a recipient with no predisposition.

Threshold model of antibody-mediated transfusion-related acute lung injury (TRALI). A specific threshold must be overcome to induce a TRALI reaction. To overcome a threshold, several factors act together: the activation status of the pulmonary neutrophils at the time of transfusion, the strength of the neutrophil-priming activity of transfused mediators (A), and the clinical status of the patient (B).

Panel 2: Clinical characteristics of transfusion-related acute lung injury (TRALI) and transfusion-associated circulatory overload (TACO)

TRALI
• Dyspnea
• Fever
• Usually hypotension
• Hypoxia
• Leukopenia
• Thrombocytopenia
• Pulmonary edema on chest x-ray
• Normal left ventricular function*
• Normal pulmonary artery occlusion pressure

TACO
• Dyspnea
• Usually hypertension
• Hypoxia
• Pulmonary edema on chest radiographs
• Normal or decreased left ventricular function
• Increased pulmonary artery occlusion pressure
• Raised brain natriuretic peptide

Restrictive transfusion policy

The most effective prevention is a restrictive transfusion strategy. In a randomised clinical trial in critically ill patients, a restrictive transfusion policy for red blood cells was associated with a decrease in incidence of acute lung injury compared with a liberal strategy (7·7% vs 11·4%), suggesting that some of these patients might have had TRALI. The restrictive threshold was well tolerated and has greatly helped in guidance of red blood cell transfusion in the intensive-care unit.

Patient-tailored transfusion policy

Transfusion cannot be avoided altogether. A multivariate analysis in patients in intensive care showed that patient related risk factors contributed more to the onset of TRALI than did transfusion-related risk factors, suggesting that development of a TRALI reaction is dependent more on host factors then on factors in the blood product. Therefore, a patient-tailored approach aimed at reducing TRALI risk factors could be effective to alleviate the risk of TRALI.

Despite limitations of diagnostic tests, TRALI incidence seems to be high in at-risk patient populations. Therefore, TRALI is an underestimated health-care problem. Preventive measures, such as mainly male donor strategies, have been successful in reducing risk of TRALI. Identification of risk factors further improves the risk–benefit assessment of a blood transfusion. Efforts to further decrease the risk of TRALI needs increased awareness of this syndrome among physicians.

 

Transfusion-related acute lung injury: Current understanding and preventive strategies

A.P.J. Vlaar
Transfusion Clinique et Biologique 19 (2012) 117–124
http://dx.doi.org/10.1016/j.tracli.2012.03.001

Transfusion-related acute lung injury (TRALI) is the most serious complication of transfusion medicine. TRALI is defined as the onset of acute hypoxia within 6 hours of a blood transfusion in the absence of hydrostatic pulmonary edema. The past decades have resulted in a better understanding of the pathogenesis of this potentially life-threating syndrome. The present notion is that the onset of TRALI follows a threshold model in which both patient and transfusion factors are essential. The transfusion factors can be divided into immune and non-immune mediated TRALI. Immune-mediated TRALI is caused by the passive transfer of human neutrophil antibodies (HNA) or human leukocyte antibodies (HLA) present in the blood product reacting with a matching antigen in the recipient. Non-immune mediated TRALI is caused by the transfusion of stored cell-containing blood products. Although the mechanisms behind immune-mediated TRALI are reasonably well understood, this is not the case for non-immune mediated TRALI. The increased understanding of pathways involved in the onset of immune-mediated TRALI has led to the design of preventive strategies. Preventive strategies are aimed at reducing the risk to exposure of HLA and HNA to the recipient of the transfusion. These strategies include exclusion of “at risk” donors and pooling of high plasma volume products and have shown to reduce the TRALI incidence effectively.

Studies show that, in at risk patient populations, up to 8% of transfused patients may develop TRALI. Since the syndrome TRALI has been recognized, evidence on the pathogenesis of TRALI has been accumulating. The present notion is that the onset of TRALI follows a threshold model in which both patient and transfusion factors are essential in the development of TRALI. The transfusion factors can be divided into immune and non-immune mediated TRALI. Immune-mediated TRALI is caused by the passive transfer of human neutrophil antibodies (HNA) or human leukocyte antibodies (HLA) present in the blood product, reacting with a matching antigen in the recipient. Non-immune mediated TRALI is caused by the transfusion of stored cell-containing blood products. In recent years, many countries have successfully implemented preventive strategies resulting in a decrease of the incidence of TRALI.

Definition of transfusion-related acute lung injury (TRALI).

  • Acute onset within 6 hours after a blood transfusion
  • PaO2/FiO2 < 300 mmHg
  • Bilateral infiltrative changes on the chest X-ray
  • No sign of hydrostatic pulmonary edema (PAOP < 18 mmHg or CVP < 15 mmHg)
  • No other risk factor for acute lung injury present

Possible TRALI

  • Other risk factor for acute lung injury present

PAOP: pulmonary arterial occlusion pressure; CVP: central venous pressure

The first landmark report creating the basis for the understanding of the pathogenesis of TRALI was published by Popovsky et al. in 1983. They provided evidence on the association between the presence of leucocyte antibodies in the donor serum and onset of acute lung injury in the recipient of the transfusion. It was also recognized that multiparous blood donors whose plasma contained these antibodies represented a potential transfusion hazard. It was this research group that was the first to identify TRALI as a distinct clinical entity. Subsequently, many other authors reported on the association between the presence of HLA or HNA antibodies in donor blood and the onset of TRALI in the recipient.

Although the role of transfused blood donor HLA and HNA antibodies was widely accepted to be involved in the onset of TRALI, not all cases could be explained by this theory. A significant part of reported TRALI cases have no detectable antibodies. Also, many antibody-containing blood products fail to produce TRALI.

The alternative hypothesis proposed by the group of Silliman posed that TRALI is a “two hit” event. The “first hit” is the underlying condition of the patient, resulting in priming of the pulmonary neutrophil. The “second hit” is the transfusion of a blood product causing activation of the neutrophils in the pulmonary compartment, causing pulmonary edema finally resulting in TRALI. The transfusion factors causing the “second hit” are divided in two groups; immune and non-immune mediated TRALI.

The “second hit” is the transfusion itself and is either immune or non-immune mediated TRALI. The mechanisms behind immune-mediated TRALI are widely accepted and proven in both pre-clinical and clinical studies.  The mechanisms involved in non-immune mediated TRALI are less clear.

The role of stored cell-containing blood products in the onset of non-immune TRALI has extensively been studied in preclinical and clinical studies. Although most of the pre-clinical studies find a positive correlation between the transfusion of stored cell-containing blood products in the presence of a “first hit” and the onset of TRALI, the mechanism behind the onset is controversial.

TRALI management consists mainly of preventing future adverse reactions and providing proper incidence estimates. All suspected TRALI cases should be reported to the blood bank for immunologic work-up as it is impossible to distinguish immune-mediated TRALI from non-immune mediated TRALI at bedside. Immunologic work-up includes testing of incompatibility by cross-matching donor plasma against recipient’s leucocytes. A donor with antibodies which are incompatible with the patient is excluded from further donation of blood for transfusion products. Furthermore, it is important to stress that the absence of a positive serologic work-up does not exclude the diagnosis of TRALI. TRALI is a clinical diagnosis and the immunologic work-up can be supportive but is not part of the diagnosis of TRALI. the two-event hypothesis and threshold hypothesis do not exclude the role of antibodies in the occurrence of TRALI in the presence of an inflammatory condition. Thus any patient fulfilling the TRALI definition (including possible TRALI) should be reported to the blood bank for an immunologic work-up of the recipient and the implicated donors on the presence of HLA and HNA antibodies.

Prevention of immune-mediated TRALI is achieved by exclusion of donors proven to have HLA or HNA antibodies in their plasma present or donors “at risk” to have these antibodies present.

  1. Exclusion of HLA or HNA positive donors
  2. Exclusion of donors “at risk” of being HLA or HNA positive
    Female donors – more specifically, multiparous donors
  3. Testing donors for HLA or HNA antibodies
  4. Multiple plasma pooling
    solvent/detergent plasma is produced from multiple donations, leading to an at least 500-fold dilution of a single plasma unit;
    neither HNA nor HLA antibodies are detectable in solvent/detergent fresh frozen plasma.
  5. To prevent non-immune mediated TRALI, the use of fresh blood only has been suggested

Strategies to prevent the onset of TRALI include the exclusion of female plasma donors and the pooling of plasma products. These strategies have already been implemented in some countries resulting in a reduction of the incidence of TRALI.
Transfusion-related immunomodulation (TRIM): An update

Eleftherios C. Vamvakas, Morris A. Blajchman
Blood Reviews (2007) 21, 327–348
http://dx.doi.org:/10.1016/j.blre.2007.07.003

Allogeneic blood transfusion (ABT)-related immunomodulation (TRIM) encompasses the laboratory immune aberrations that occur after ABT and their established or purported clinical effects. TRIM is a real biologic phenomenon resulting in at least one established beneficial clinical effect in humans, but the existence of deleterious clinical TRIM effects has not yet been confirmed. Initially, TRIM encompassed effects attributable to ABT by immunomodulatory mechanisms (e.g., cancer recurrence, postoperative infection, or virus activation). More recently, TRIM has also included effects attributable to ABT by pro-inflammatory mechanisms (e.g., multiple-organ failure or mortality). TRIM effects may be mediated by: (1) allogeneic mononuclear cells; (2) white-blood-cell (WBC)-derived soluble mediators; and/or (3) soluble HLA peptides circulating in allogeneic plasma. This review categorizes the available randomized controlled trials based on the inference(s) that they permit about possible mediator(s) of TRIM, and examines the strength of the evidence available for relying on WBC reduction or autologous transfusion to prevent TRIM effects.

Allogeneic blood transfusion (ABT) may either cause alloimmunization or induce tolerance in recipients. ABTs introduce a multitude of foreign antigens into the recipient, including HLA-DR antigens found on the donor’s dendritic antigen presenting cells (APCs). The presence or absence of recipient HLA-DR antigens on the donor’s white blood cells (WBCs) plays a decisive role as to whether alloimmunization or immune suppression will ensue following ABT. In general, allogeneic transfusions sharing at least one HLA-DR antigen with the recipient induce tolerance, while fully HLA-DR-mismatched transfusions lead to alloimmunization.

In addition to the degree of HLA-DR compatibility between donor and recipient, the immunogenicity of cellular or soluble HLA antigens associated with transfused blood components depends on the viability of the donor dendritic APCs and the presence of co-stimulatory signals for the presentation of the donor antigens to the recipient’s T cells. Nonviable APCs and/or the absence of the requisite co-stimulatory signals result in T-cell unreponsiveness.  Thus, when a multitude of antigens is introduced into the host by an ABT, the host response to some of these antigens is often decreased, and immune tolerance ensues. ABT has been shown to cause decreased helper T-cell count, decreased helper/suppressor T-lymphocyte ratio, decreased lymphocyte response to mitogens, decreased natural killer (NK) cell function, reduction in delayed-type hypersensitivity, defective antigen presentation, suppression of lymphocyte blastogenesis, decreased cytokine (IL-2, interferon-c) production, decreased monocyte/macrophage phagocytic function, and increased production of antiidiotypic and anticlonotypic antibodies.

All these laboratory immune aberrations that indicate immune suppression and occur in transfused patients could potentially be associated with clinically-manifest ABT effects. Thus a variety of beneficial or deleterious clinical effects, potentially attributable to ABT-related immunosuppression, have been described over the last 30 years. The constellation of all such ABT-associated laboratory and clinical findings is known as ABT-related immunomodulation (TRIM). Initially, TRIM encompassed effects attributable to ABT by means of immunologic mechanisms only; however more recently, the term has been used more broadly, to encompass additional effects that could be related to ABT by means of ‘‘proinflammatory’’ rather than ‘‘immunomodulatory’’ mechanisms.

Over 30 years ago, it was reported that pre-transplant ABTs could improve renal-allograft survival in patients who had undergone renal transplantation.  This beneficial immunosuppressive effect of ABT has been confirmed by animal data, observational clinical studies, and clinical experience worldwide, although it has not been proven in randomized controlled trials (RCTs). Before the advent of the AIDS pandemic, it had become standard policy in many renal units to deliberately expose patients on transplant waiting lists to one or more red blood cell (RBC) transfusions.

All the available data considered together indicate that TRIM is most likely a real biologic phenomenon, which results in at least one established beneficial clinical effect in humans, although the available evidence has not yet confirmed  the existence and/or magnitude of the deleterious clinical TRIM effects. In fact, the debate over the existence of such deleterious clinical TRIM effects has been long and sometimes acrimonious.

Many studies tended to indicate that patients receiving perioperative transfusion (compared with those not needing transfusion) almost always had a higher risk of developing postoperative bacterial infection. The studies also indicated that patients receiving ABT differed from those not receiving a transfusion in several prognostic factors that predisposed to adverse clinical outcomes.

The specific constituent(s) of allogeneic blood that mediate(s) either or both the immunomodulatory and the pro-inflammatory effect(s) of ABT remain
(s) unknown, and the published literature suggests that these TRIM effects
may be mediated by: (1) allogeneic mononuclear cells; (2) soluble biologic response modifiers released in a time dependent manner from WBC granules or membranes into the supernatant fluid of RBC or platelet concentrates
during storage; and/or  (3) soluble HLA class I peptides that circulate in allogeneic plasma. If each of these mediators do cause TRIM effects, ABT effects mediated by allogeneic mononuclear cells would be expected to be preventable by WBC reduction (performed either before or after storage of cellular blood components), as well as by autologous transfusion. The ABT effects mediated by soluble HLA peptides circulating in allogeneic plasma would be expected to be preventable only by autologous transfusion.

BENEFICIAL TRIM EFFECTS

  1. Enhanced survival of renal allografts
  2. Reduced recurrence rate of Crohn’s disease

DELETERIOUS

  1. Increased recurrence rate of resected malignancies
  2. Increased incidence of postoperative bacterial infections
  3. Activation of endogenous CMV or HIV infection
  4. Increased short-term (up to 3-month) mortality

Possible mechanisms and mediators of TRIM effects

Although the mechanisms of TRIM have been debated extensively, the exact mechanism(s) of this phenomenon has yet to be elucidated. A number of putative mechanisms have been postulated. The three major mechanisms accounting for much of the experimental data include:

  • clonal deletion,
  • induction of anergy, and
  • immune suppression.

Conceptually, clonal deletion refers to the inactivation and removal of alloreactive lymphocytes that would, for example, cause the rejection of an allograft; anergy implies immunologic nonresponsiveness; and immune suppression suggests that the responding cell is being inhibited of doing so by a cellular mechanism or by a cytokine. Antiidiotypic antibodies, which are predominantly of the VH6 gene family, have also been demonstrated in the sera of ABT recipients and in patients with long-term functioning renal allografts.

To date, no RCT has enrolled patients with sarcomas—tumors whose growth is stimulated by TGF-β—or patients with tumors for which the immune response plays a major role. (These would include skin tumors—such as melanomas, keratoacanthomas, squamous and basal-cell carcinomas—and certain virus-induced tumors—notably Kaposi’s sarcoma and certain lymphomas.) Instead, the 3 available RCTs of ABT and cancer recurrence enrolled patients with colorectal cancer—a tumor that is not sufficiently antigenic to render an impairment of host immunity capable of facilitating tumor growth, and a tumor whose cells have not been shown to be stimulated by TGF-β.

Fig not shown. Randomized controlled trials (RCTs) investigating the association of WBC-containing allogeneic blood transfusion (ABT) with cancer recurrence. For each RCT, the figure shows the odds ratio (OR) of cancer recurrence in recipients of non-WBC-reduced allogeneic versus autologous or WBC-reduced allogeneic RBCs, as calculated from an intention-to-treat analysis. A deleterious effect of ABT (and thus a benefit from autologous transfusion or WBC reduction) exists when the OR is greater than 1 as well as statistically significant. (In the figure, each OR is surrounded by its 95% confidence interval [CI]; if the 95% CI of the OR includes the null value of 1, the TRIM effect is not statistically significant [p > 0.05]).

Fig not shown. Randomized controlled trials (RCTs) investigating the association of WBC-containing allogeneic blood transfusions with postoperative infection (n = 17). For each RCT, the figure shows the odds ratio (OR) of postoperative infection in recipients of non-WBC reduced allogeneic versus autologous or WBC-reduced allogeneic RBCs, as calculated from an intention-to-treat analysis. A deleterious effect of ABT (and thus a benefit from autologous transfusion or WBC reduction) exists when the OR is greater than 1 as well as statistically significant. (In the figure, each OR is surrounded by its 95% confidence interval [CI]; if the 95% CI of the OR includes the null value of 1, the TRIM effect is not statistically significant [p > 0.05]).

The totality of the evidence from RCTs does not demonstrate a TRIM effect manifest across all clinical settings and transfused RBC products. Instead, WBC-containing ABT is associated with an increased risk of short-term (up to 3-month post transfusion) mortality from all causes combined specifically in cardiac surgery. The additional deleterious TRIM effect detected by the latest meta-analysis (i.e., the effect on postoperative infection prevented by poststorage filtration) contradicts current theories about the pathogenesis of TRIM, because it is not accompanied by a similar or larger effect prevented by prestorage filtration.

Thus, only in cardiac surgery (Fig. 5 – not shown) are the findings of RCTs pertaining to a deleterious TRIM effect consistent. Even in this setting, however, the reasons for the excess deaths attributed to WBC containing ABT remain elusive. The initial hypothesis suggested that WBC-containing ABT may predispose to MOF which, in turn, may predispose to mortality. However, hitherto, no cardiac-surgery RCT has demonstrated an association between WBC-containing ABT and MOF, and no other cause of death specifically attributed to WBC-containing ABT has been proposed.

The TRIM effect seen in cardiac surgery deserves further study to pinpoint the cause(s) of the excess deaths, but-now that the majority of transfusions in Western Europe and North America are WBC reduced- the undertaking of further RCTs comparing recipients of non-WBC-reduced versus WBC reduced allogeneic RBCs in cardiac surgery is unlikely. For countries that have not yet converted to universal WBC reduction, whether to opt for WBC reduction of all cellular blood components transfused in cardiac surgery-in the absence of information on the specific cause(s) of death ascribed to WBC-containing ABT-is a policy decision that will have to be made based on the hitherto available data.

 

Regulation of alveolar fluid clearance and ENaC expression in lung by exogenous angiotensin II

Jia Denga, Dao-xin Wanga, Wang Deng, Chang-yi Li, Jin Tong, Hilary Ma
Respiratory Physiology & Neurobiology 181 (2012) 53– 61
http://dx.doi.org:/10.1016/j.resp.2011.11.009

Angiotensin II (Ang II) has been demonstrated as a pro-inflammatory effect in acute lung injury, but studies of the effect of Ang II on the formation of pulmonary edema and alveolar filling remains unclear. Therefore, in this study the regulation of alveolar fluid clearance (AFC) and the expression of epithelial sodium channel (ENaC) by exogenous Ang II was verified. SD rats were anesthetized and were given Ang II with increasing doses (1, 10 and 100 [1]g/kg per min) via osmotic minipumps, whereas control rats received only saline vehicle. AT1 receptor antagonist ZD7155 (10 mg/kg) and inhibitor of cAMP degeneration rolipram (1 mg/kg) were injected intraperitoneally 30 min before administration of Ang II. The lungs were isolated for measurement of alveolar fluid clearance. The mRNA and protein expression of ENaC were detected by RT-PCR and Western blot. Exposure to higher doses of Ang II reduced AFC in a dose-dependent manner and resulted in a non-coordinate regulation of α-ENaC vs the regulation of β- and ϒ-ENaC, however Ang II type 1 (AT1) receptor antagonist ZD7155 prevented the Ang II-induced inhibition of fluid clearance and dysregulation of ENaC expression. In addition, exposure to inhibitor of cAMP degradation rolipram blunted the Ang II-induced inhibition of fluid clearance. These results indicate that through activation of AT1 receptor, exogenous Ang II promotes pulmonary edema and alveolar filling by inhibition of alveolar fluid clearance via downregulation of cAMP level and dysregulation of ENaC expression.

Effects of angiotensin II (Ang II) receptor antagonists and rolipram  on AFC

Effects of angiotensin II (Ang II) receptor antagonists and rolipram on AFC

Effects of angiotensin II (Ang II) receptor antagonists and rolipram on rat alveolar fluid clearance (AFC). Then AFC was measured 1 h after fluid instillation (4 mL/kg). Amiloride (100 [1]M), Ang II (10−7 M), ZD7155 (10−6 M), and rolipram (10−5 M) were added to the instillate as indicated (n = 10 per group). Mean values ± SEM. p < 0.01 vs control. p < 0.01 vs Ang II + ZD7155.
p < 0.05 vs amiloride. p < 0.05 vs Ang II.

Effects of angiotensin II (Ang II) on cyclic adenosine monophosphate (cAMP)

Effects of angiotensin II (Ang II) on cyclic adenosine monophosphate (cAMP)

Effects of angiotensin II (Ang II) on cyclic adenosine monophosphate (cAMP) concentration in lung. Rats were given saline or Ang II (1, 10 and 100 µg/kg per min) for 6 h, and cAMP in lung was determined by RIA (n = 30 per group). Mean values ± SEM. p < 0.01 vs control. p < 0.05 vs 10 µg/kg Ang II.

Histological examination of lung

Histological examination of lung

Histological examination of lung. Rats were given saline or Ang II (10 µg/kg per min) by osmotic minipump for 6 h. ZD7155 (10 mg/kg) was injected intraperitoneally 30 min before administration of Ang II. Shown are representative lung specimens obtained from the control (A), Ang II (B) and Ang II + ZD7155 (C) groups. All photographs are at 100× magnification. Interstitial edema and inflammatory cell infiltration were seen in Ang II group, but reduced in Ang II + ZD7155 group.
The present results demonstrate that Ang II infusion is associated with pulmonary edema and alveolar filling. Three important findings were observed:

(1) high doses of Ang II led to reduction of alveolar fluid clearance, and this effect was blunted by an AT1 receptor antagonist.
(2) Ang II infusion increased the abundance of α-ENaC, whereas decreased the abundance ofβ and ϒ-ENaC, and these effects were reversed in response to an AT1 receptor antagonist.
(3) Ang II infusion decreased cAMP concentration in lung tissue, and an inhibitor of cAMP degradation prevented inhibition of alveolar fluid clearance by Ang II, but had no effect on the dysregulation of ENaC.

Our data indicate that Ang II results in pulmonary edema by inhibition of alveolar fluid clearance via down-regulation of cellular cAMP level and dysregulation of the abundance of ENaC, whereas these effects are prevented by an AT1 receptor antagonist.

The renin-angiotensin system is a major regulator of body fluid and sodium balance, predominantly through the actions of its main effector Ang II. Several previous experimental studies demonstrated that plasma Ang II levels vary in both physiological and pathological conditions. In the kidney, Ang II added to the peritubular perfusion has a biphasic action with stimulation of sodium reabsorption at low doses (10−12–10−10M) and inhibition at high doses (10−7–10−6M) (Harris and Young, 1977). In vitro, Ang II also exerts a dose-dependent dual action on intestinal absorption (Levens, 1985). The evidence shows that the effect of Ang II on sodium and water absorption is dose-dependent. Our results showed that low intravenous doses of Ang II (<1 µg/kg per min) had no effect on alveolar fluid clearance which represents the sodium and water reabsorption in alveoli. However, with high intravenous doses, Ang II decreased alveolar fluid clearance. This finding suggests that the effect of Ang II on fluid absorption in lung is also dose-dependent.

 

Rat models of acute lung injury: Exhaled nitric oxide as a sensitive,noninvasive real-time biomarker of prognosis and efficacy of intervention

Fangfang Liu, Wenli Lib, Jürgen Pauluhn, Hubert Trübel, Chen Wang
Toxicology 310 (2013) 104– 114
http://dx.doi.org/10.1016/j.tox.2013.05.016

Exhaled nitric oxide (eNO) has received increased attention in clinical settings because this technique is easy to use with instant readout. However, despite the simplicity of eNO in humans, this endpoint has not frequently been used in experimental rat models of septic (endotoxemia) or irritant acute lung injury (ALI). The focus of this study is to adapt this method to rats for studying ALI-related lung disease and whether it can serve as instant, non-invasive biomarker of ALI to study lung toxicity and pharmacological efficacy. Measurements were made in a dynamic flow of sheath air containing the exhaled breath from spontaneously breathing, conscious rats placed into a head-out volume plethysmograph. The quantity of eNO in exhaled breath was adjusted (normalized) to the physiological variables (breathing frequency, concentration of exhaled carbon dioxide) mirroring pulmonary perfusion and ventilation. eNO was examined on the instillation/inhalation exposure day and first post-exposure day in Wistar rats intratracheally instilled with lipopolysaccharide (LPS) or single inhalation exposure to chlorine or phosgene gas. eNO was also examined in a Brown Norway rat asthma model using the asthmagen toluene diisocyanate (TDI). The diagnostic sensitivity of adjusted eNO was superior to the measurements not accounting forthe normalization of physiological variables. In all bioassays – whether septic, airway or alveolar irritant or allergic, the adjusted eNO was significantly increased when compared to the concurrent control. The maximum increase of the adjusted eNO occurred following exposure to the airway irritant chlorine. The specificity of adjustment was experimentally verified by decreased eNO following inhalation dosing ofthe non-selective nitric oxide synthase inhibitor amoni-guanidine. In summary, the diagnostic sensitivity of eNO can readily be applied to spontaneously breathing, conscious rats without any intervention or anesthesia. Measurements are definitely improved by accounting for the disease-related changes inexhaled CO2and breathing frequency. Accordingly, adjusted eNO appears to be a promising methodological improvement for utilizing eNO in inhalation toxicology and pharmacological disease models
with fewer animals.

 

Role of p38 MAP Kinase in the Development of Acute Lung Injury

J Arcaroli, Ho-Kee Yum, J Kupfner, JS Park, Kuang-Yao Yang, and E Abraham
Clinical Immunology 2001; 101(2):211–219
http://dx.doi.org:/10.1006/clim.2001.5108

Acute lung injury (ALI) is characterized by an intense pulmonary inflammatory response, in which neutrophils play a central role. The p38 mitogen-activated protein kinase pathway is involved in the regulation of stress-induced cellular functions and appears to be important in modulating neutrophil activation, particularly in response to endotoxin. Although p38 has potent effects on neutrophil functions under in vitro conditions, there is relatively little information concerning the role of p38 in affecting neutrophil driven inflammatory responses in vivo. To examine this issue, we treated mice with the p38 inhibitor SB203580 and then examined parameters of neutrophil activation and acute lung injury after hemorrhage or endotoxemia. Although p38 was activated in lung neutrophils after hemorrhage or endotoxemia, inhibition of p38 did not decrease neutrophil accumulation in the lungs or the development of lung edema under these conditions. Similarly, the increased production of proinflammatory cytokines and activation of NF-kB in lung neutrophils induced by hemorrhage or endotoxemia was not diminished by p38 inhibition. These results indicate that p38 does not have a central role
in the development of ALI after either hemorrhage or endotoxemia.

 

The coagulation system and pulmonary endothelial function in acute lung injury

James H. Finigan
Microvascular Research 77 (2009) 35–38
http://dx.doi.org:/10.1016/j.mvr.2008.09.002

Acute lung injury (ALI) is a disease marked by diffuse endothelial injury and increased capillary permeability. The coagulation system is a major participant in ALI and activation of coagulation is both a consequence and contributor to ongoing lung injury. Increased coagulation and depressed fibrinolysis result in diffuse alveolar fibrin deposition which serves to amplify pulmonary inflammation. In addition, existing evidence demonstrates a direct role for different components of coagulation on vascular endothelial barrier function. In particular, the pro-coagulant protein thrombin disrupts the endothelial actin cytoskeleton resulting in increased endothelial leak. In contrast, the anti-coagulant activated protein C (APC) confers a barrier protective actin configuration and enhances the vascular barrier in vitro and in vivo. However, recent studies suggest a complex landscape with receptor cross-talk, temporal heterogeneity and pro-coagulant/anticoagulant protein interactions. In this article, the major signaling pathways governing endothelial permeability in lung injury are reviewed with a particular focus on the role that endothelial proteins, such as thrombin and APC, which play on the vascular barrier function.

Acute lung injury (ALI) is a devastating illness with an annual incidence of approximately 200,000 and a mortality of 40%. Most commonly seen in the setting of sepsis, ALI is a complex inflammatory syndrome marked by increased vascular permeability resulting in tissue edema and organ dysfunction. The vascular endothelium is a key target and critical participant in the pathogenesis of sepsis-induced organ dysfunction and disruption of the endothelial barrier is central to the pathophysiology of both sepsis and ALI. Sepsis and acute lung injury (ALI) are syndromes marked by diffuse inflammation with a key feature being endothelial cell barrier disruption and increased vascular permeability resulting in widespread organ dysfunction. The endothelial cytoskeleton has been identified as a critical regulator of vascular barrier integrity with a current model of endothelial barrier regulation suggesting a balance between barrier-disrupting cellular contractile forces and barrier-protective cell–cell and cell–matrix forces. These competing forces exert their opposing effects via manipulation of the actin-based endothelial cytoskeleton and associated endothelial regulatory proteins. Endothelial cells generate tension via an actomyosin motor, and focally distributed changes in tension/relaxation can be accomplished by spatially-defined regulation of the phosphorylation of the regulatory 20 kDa myosin light chain (MLC) catalyzed by the Ca2+/calmodulin (CaM)-dependent enzyme myosin light chain kinase (MLCK).

Thrombin is the proto-typical coagulation protein with direct effects on the endothelial barrier via alterations in the cytoskeleton. In the coagulation cascade, thrombin converts fibrinogen to fibrin in the final step of thrombus formation and also activated platelets. In addition, this multifunctional protease is present at sites of vascular inflammation and induces barrier dysfunction. Through its receptor, protease-activated receptor-1 (PAR1), thrombin initiates a series of events which includes MLC phosphorylation, dramatic cytoskeletal reorganization and stress fiber formation, increased cellular contractility, paracellular gap formation, and enhanced fluid and protein transport. Similarly, thrombin exposure results in increased pulmonary edema in vivo, a finding which is also seen after treatment with a PAR1 activating peptide and attenuated in PAR1 knockout mice.

Disruptions in the coagulation system have long been recognized to be an integral part of inflammation, sepsis and ALI. In 1969, Saldeen demonstrated that thrombin infusion produced canine respiratory insufficiency which was linked pathologically to emboli in the pulmonary microcirculation, a condition he labeled the “Microembolism Syndrome” (Saldeen, 1979). Elemental to the pathophysiology of sepsis and ALI is a shift towards a pro-coagulant state. Bronchoalveolar (BAL) fluid from patients with ALI reflects this increase in procoagulant activity with elevated levels of fibrinopeptide A, factor VII and d-dimer. Concomitantly, there is a decrease in fibrinolytic activity, as shown by depressed BAL levels of urokinase and increased levels of the fibrinolysis inhibitors plasminogen activator inhibitor (PAI) and α2-antiplasmin.

Given that APC is a vascular endothelial protein which interacts with other coagulation proteins such as thrombin, it seems logical that it might have an effect on endothelial integrity. In cultured human pulmonary endothelial cells, while thrombin results in decreased electrical resistance, a reflection of increased permeability, pre- or post-exposure to physiologic concentrations of APC significantly attenuates this thrombin-induced drop in resistance. These APC-mediated alterations in barrier function are associated with MLC phosphorylation as well as activation of the endothelial protein Rac, and cytoskeletal re-arrangement in a barrier protective configuration all findings very reminiscent of the barrier protective signaling induced by the bioactive lipid, S1P. Interestingly, APC appears to activate sphingosine kinase and mediate its barrier protective effects through PI3 kinase and AKT-dependent ligation of the S1P receptor, S1P1. Moreover, the endothelial barrier-protective effects of APC have been observed in other tissues including brain and kidney. The barrier protection in these beds appears independent of any anti-coagulant effect of APC and is associated with decreased endothelial apoptosis.

Recently, the endothelial protein C receptor (EPCR) has been identified as a crucial participant in the protein C pathway. Structurally similar to the major histocompatibility class I/CD1 family of molecules, EPCR binds protein C, presenting it to the thrombin/TM complex, thereby increasing the activation of protein C by ∼20 fold. Importantly, APC can also bind EPCR, and while the bound form of APC loses its extra-cellular anti-coagulant activity, increasing evidence indicates that much, if not all, of APC intra-cellular signaling requires EPCR. APC-mediated increases in endothelial phosphor-MLC and activated Rac are all EPCR-dependent and APC-induced endothelial barrier protection requires ligation of EPCR.

Sepsis and ALI are significant causes of morbidity and mortality in the intensive care unit and are marked by zealous activation of the coagulation system. While this could conceivably confer certain benefits, such as enclosing and spatially controlling an infection, it is clear that this pro-coagulant environment participates in the pathophysiology of ALI, particularly via exacerbating endothelial damage and augmenting endothelial permeability. However, the biology of coagulation in ALI is incompletely understood and trials of new therapies specifically targeting coagulation in patients with ALI have been disappointing. Despite this, recent advances in the knowledge of the dynamic interplay between inflammation and coagulation in ALI as well as endothelial receptor-ligand binding and receptor cross talk have stimulated promising research and identified novel therapeutic targets for patients with ALI.

 

Phosphatidylserine-expressing cell by-products in transfusion: A pro-inflammatory or an anti-inflammatory effect?

  1. Saas, F. Angelot, L. Bardiaux, E. Seilles, F. Garnache-Ottou, S. Perruche
    Transfusion Clinique et Biologique 19 (2012) 90–97
    http://dx.doi.org/10.1016/j.tracli.2012.02.002

Labile blood products contain phosphatidylserine-expressing cell dusts, including apoptotic cells and microparticles. These cell by-products are produced during blood product process or storage and derived from the cells of interest that exert a therapeutic effect (red blood cells or platelets). Alternatively, phosphatidylserine-expressing cell dusts may also derived from contaminating cells, such as leukocytes, or may be already present in plasma, such as platelet-derived microparticles. These cell by-products present in labile blood products can be responsible for transfusion induced immunomodulation leading to either transfusion-related acute lung injury (TRALI) or increased occurrence of post-transfusion infections or cancer relapse. In this review, we report data from the literature and our laboratory dealing with interactions between antigen-presenting cells and phosphatidylserine-expressing cell dusts, including apoptotic leukocytes and blood cell-derived microparticles. Then, we discuss how these phosphatidylserine-expressing cell by-products may influence transfusion.

Potential consequences of phosphatidylserine-expressing cell by-products in transfusion

Potential consequences of phosphatidylserine-expressing cell by-products in transfusion

Potential consequences of phosphatidylserine-expressing cell by-products in transfusion. Interactions of phosphatidylserine-expressing cell dusts (apoptotic cells or microparticles) may lead to antigen-presenting cell activation or inhibition. Antigen-presenting cell activation may trigger inflammation and be involved in transfusion-related acute lung injury (TRALI), while antigen-presenting cell inhibition may exert transient immunosuppression or tolerance. Blood product process or storage may influence the generation of phosphatidylserine-expressing cell dusts. PtdSer: phosphatidylserine; APC: antigen-presenting cell.

Several publications report the presence of phosphatidylserine-expressing cell by-products in blood products. These cell by-products may be generated during the blood product process, such as filtration, or during storage (either cold storage for red blood cells or between 20–24 ◦C for platelets). Alternatively, they may be limited by filtration. Phosphatidylserine-expressing cell by-products can be apoptotic cells. Apoptotic cells have been found in different blood products: red blood cell units and platelet concentrates. These apoptotic cells correspond to dying cells of interest: red blood cells or platelets, both enucleated cells that can undergo apoptosis.

Immunomodulatory effects of apoptotic leukocytes

Immunomodulatory effects of apoptotic leukocytes

Immunomodulatory effects of apoptotic leukocytes. Early during the apoptotic program, phosphatidylserine-exposure occurs leading to apoptotic cell removal by macrophages or conventional dendritic cells. This uptake by antigen-presenting cells induces the production of anti-inflammatory factors and concomitantly inhibits the synthesis of inflammatory cytokines. These antigen-presenting cells are refractory to TLR activation. This leads to a transient immunosuppressive microenvironment. If antigen-presenting cells from this microenvironment migrate to secondary lymphoid organs, naive T cells are converted into inducible regulatory T cells. This leads to tolerance against apoptotic cell-derived antigens. M[1]: macrophage; cDC: conventional dendritic cells; PtdSer: phosphatidylserine; Treg: regulatory T cells; Th1: helper T cells; HGF: hepatocyte growth factor; IL-: interleukin; NO: nitrite oxide; PGE-2: prostaglandin-E2; TGF: transforming growth factor; TNF: tumor necrosis factor; TLR: Toll-like receptor.

Implication of phosphatidylserine in the inhibition of both inflammation and specific immune responses has been further demonstrated using  phosphatidylserine-expressing liposomes and is sustained by the following observations:

  • phosphatidylserine-dependent ingestion of apoptotic cells induces TGF-β secretion and resolution of lung inflammation;
  • inhibition of phosphatidylserine recognition through annexin-V enhances the immunogenicity of irradiated tumor cells in vivo;
  • masking of phosphatidylserine inhibits apoptotic cell engulfment and induces autoantibody production in mice.

Based on data from our group and Peter Henson’s group, some authors have speculated that apoptotic leukocytes present in blood products may be responsible for transfusion-related immunosuppression.

The first consequences of phosphatidylserine-expressing apoptotic cells in blood products may be a transient immunosuppression−responsible for an increase in infection rate and of cancer relapse−or tolerance induction− as observed after donor-specific transfusion − when Treg have been generated. However, apoptotic leukocytes become secondarily necrotic in the absence of phagocytes. This may certainly occur in blood product bags. Necrotic cells, through the release of damage-associated molecular patterns, may become immunogenic. The same process may occur for platelets. Necrotic platelets may represent the procoagulant form of platelets. Thus, hemostatic activation of platelets or their by-products may link thrombosis and inflammation to amplify lung microvascular damage during nonimmune TRALI.

What are the next steps to answer the question on the role of phosphatidylserine-expressing cell dusts in the modulation of immune responses after transfusion?

The next steps are to characterize or identify factors involved in the triggering of inflammation or its inhibition and produced during blood product storage or process. Several factors influence the immune responses against dying cells. We can speculate on some factors, including:

  • the number of phosphatidylserine-expressing cell byproducts contained per blood product, as the immunogenicity of apoptotic cells may be proportional to their number;
  • the occurrence of secondary necrosis and so the passive release of intracellular damage-associated molecular patterns that overpasses the inhibitory signals delivered by phosphatidylserine. One of these damage associated molecular patterns can be the heme released from stored red blood cells which signals via TLR4;
  • the size of cell by-products and especially microparticles, since these latter exert different functions according to their size. Moreover, antigen-presenting cells, such as plasmacytoid dendritic cells, respond only to lower size synthetic particles. This may explain the different responses observed between “amateur” phagocytes (plasmacytoid dendritic cells) versus professional phagocytes (conventional dendritic cells/macrophages) after incubation with microparticles. The size of cell by-products diminishes during plasma filtration, as assessed by dynamic light scattering from 101 to 464 nm in unfiltered fresh-frozen plasma versus 21 to 182 nm after 0.2 µm filtration process;
  • expression of the recently described phosphatidylserine receptors on different antigen-presenting cell subsets may also explain the different responses between plasmacytoid dendritic cells versus conventional dendritic cells/macrophages and may impact on the overall immune response.

 

Peroxisome proliferator-activated receptors and inflammation

Leonardo A. Moraes, Laura Piqueras, David Bishop-Bailey
Pharmacology & Therapeutics 110 (2006) 371 – 385
http://dx.doi.org:/10.1016/j.pharmthera.2005.08.007

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptors family. PPARs are a family of 3 ligand-activated transcription factors: PPARa (NR1C1), PPARh/y (NUC1; NR1C2), and PPARg (NR1C3). PPARα, -h/y, and -ϒ are encoded by different genes but show substantial amino acid similarity, especially within the DNA and ligand binding domains. All PPARs act as heterodimers with the 9-cis-retinoic acid receptors (retinoid X receptor; RXRs) and play important roles in the regulation of metabolic pathways, including those of lipid of biosynthesis and glucose metabolism, as well as in a variety of cell differentiation, proliferation, and apoptosis pathways. Recently, there has been a great deal of interest in the involvement of PPARs in inflammatory processes. PPAR ligands, in particular those of PPARα and PPARϒ, inhibit the activation of inflammatory gene expression and can negatively interfere with proinflammatory transcription factor signaling pathways in vascular and inflammatory cells. Furthermore, PPAR levels are differentially regulated in a variety of inflammatory disorders in man, where ligands appear to be promising new therapies.

Fig. not shown.  Structure and transcriptional activation of PPARs. (A) Generic schematic of the structure of the PPAR family of nuclear receptors. Indicated are the N–C terminal regions subdivided in to 4 domains: the A/B, N terminal domain [also called the activation function (AF)-1 domain]; C, the DNA binding domain; D, the F hinge_region; and E, the ligand binding domain (AF-2). (B) Generic scheme for the activation of a PPAR receptor as a transcription factor. PPAR activation leads to heterodimerization with RXR and an accumulation in the nucleus. Ligand activation of PPAR results in a change from a repressed binding protein complex which may contain histone deacetylases (HDAC), the nuclear receptor corepressor (NCo-R), and the silencing mediator of retinoid and thyroid signaling (SMRT) to an activation complex that may contain the histone acetylases, steroid receptor co-activator-1 (SRC-1), the PPAR binding protein (PBP), cAMP response element binding protein (CBP/p300), TATA box binding proteins, and RNA polymerase (RNA pol) III. The activated PPAR–RXR heterodimer complex binds to DNA sequences called PPAR response elements (PPRE) in target genes initiation their transcription.

Although the nature of true endogenous PPAR ligands are still not known (Bishop-Bailey & Wray, 2003), PPARs can be activated by a wide variety of F endogenous or pharmacological ligands. PPARα activators include a variety of endogenously present fatty acids, LTB4 and hydroxyeicosatetraenoic acids (HETEs), and clinically used drugs, such as the fibrates, a class of first-line drugs in the treatment of dyslipidemia. Similarly, PPARg can be activated by a number of ligands, including docosahexaenoic acid, linoleic acid, the anti-diabetic glitazones, used as insulin sensitizers, and a number of lipids, including oxidized LDL, azoyle-PAF, and eicosanoids, such as 5,8,11,14-eicosatetraynoic acid and the prostanoids PGA1, PGA2, PGD2, and its dehydration products of the PGJ series of cyclopentanones (e.g., 15 deoxy-D12,14-PGJ2). Dyslipidemia and insulin-dependent diabetes are commonly found existing together as part of the metabolic X syndrome.

Because PPARa and PPARg ligands independently are useful clinical drugs in the treatment of these respective disorders, synthetic dual PPARα/ϒ ligands have recently been developed and show a combined clinical efficacy. PPAR h/y activators include fatty acids and prostacyclin and synthetic compounds L-165,041, GW501516, compound F and L-783,483. Unlike PPARα or-ϒ, there are no PPAR h/y drugs in the clinic, although ligands are in phase II clinical trials for dyslipidemia (http://www.science.gsk.com/pipeline). Indeed, part of the challenge in determining the function of PPARh/y has been the identification and availability of new ligands with more potency and selectivity for use as pharmacological tools.

Fig. not shown. Mechanisms of the anti-inflammatory effects of PPARα. PPARα ligands inhibit the activities of NF-nB, AP-1, and T-bet within cells. In sites of local inflammation, tissue and endothelial cell activity is inhibited, and expressions of adhesion molecules (ICAM-1 and VCAM-1), pro-inflammatory cytokines (IL-1, -6, -8, -12, and TNFα), vasoactive mediators (inducible cyclo-oxygenase, inducible nitric oxide synthase, and endothelin-1; COX-2, iNOS, and ET-1), and proteases (MMP-9) are decreased. The inflammatory responses in leukocytes are also diminished. Monocyte/macrophage activity is decreased, and lipid metabolizing pathways increased, T- and B-lymphocyte proliferation and differentiation are inhibited, and T-lymphocyte and eosinophil chemotaxis reduced. Bold italic text indicates positive regulation by the PPAR, all other text indicates a negative regulation.

Fig. not shown. Mechanisms of the anti-inflammatory effects of PPAR h/y. PPAR h/y ligands inhibit the activities of NF-nB and release the suppressor BCL-6 from PPAR h/y. In sites of local inflammation, endothelial cell adhesion molecule (VCAM-1) and chemokine (MCP-1) are reduced. PPAR h/y and its endogenous ligand(s) are induced during the inflammatory response in keratinocytes, which then promotes cell survival (integrin-linked kinase—Akt pathway) and wound healing. The inflammatory responses in monocyte/ macrophages are modulated. In the absence of ligand, PPAR h/y sequesters BCL-6 and induces MCP-1, MCP-3, and IL-1h. When PPAR h/y ligand is given, BCL-6 is released and MCP-1, -3, and IL-1h levels are reduced. Bold italic text indicates positive regulation by the PPAR, all other text indicates a negative regulation.

Fig. not shown. Mechanisms of the anti-inflammatory effects of PPARg. PPARg ligands can inhibit the activities of NF-nB, AP-1, STAT-1, N-FAT, Erg-1, Jun, and GATA-3 within cells. In sites of local inflammation, tissue and endothelial cell activity is inhibited, and expression of adhesion molecules (ICAM-1), proinflammatory cytokines (IL-8, -12, and TNFα), chemokines (MCP-1, MCP-3, IP-10, Mig, and I-TAC), vasoactive mediators (inducible nitric oxide synthase and endothelin-1; iNOS and ET-1), and proteases (MMP-9) are decreased. The inflammatory responses in leukocytes are also diminished. Monocyte/ macrophage activity is decreased, T- and B-lymphocyte proliferation and differentiation are inhibited, and T-lymphocyte and eosinophil chemotaxis reduced. Platelet activity is inhibited and dendritic cell production of IL-12, and expression of CCL3, CCL5, and CD80 is reduced, so pro-inflammatory TH1 lymphocytes maturation is inhibited. Bold italic text indicates positive regulation by the PPAR, all other text indicates a negative regulation.

The PPARs are one of the most intensely studied members of the nuclear receptor gene family, and since their initial discovery just over decade ago, the PPARs have attracted an increasing amount of experimental and clinical research by investigators from different scientific areas. PPARs through their central roles in regulating energy homeostasis regulate physiological function in many cell types, tissues, and organ systems. Many disease states from carcinogenesis to inflammation have been linked to abnormalities in the function of PPAR-regulated transcription factors. PPARs are expressed or regulate pathophysiology of diverse human disorders including atherosclerosis, inflammation, obesity, diabetes, and the immune response. PPARs have beneficial effects in many inflammatory conditions, where they regulate cytokine production, adhesion molecule expression, fibrinolysis cell proliferation, apoptosis, and differentiation. Further studies and development of novel PPAR ligands and their selective modulators may lead to novel therapeutic agents in the many conditions associated with inflammatory processes.

 

Regulators of endothelial and epithelial barrier integrity and function in acute lung injury

Rudolf Lucas, Alexander D. Verin, Stephen M. Black, John D. Catravas
Biochemical Pharmacology 77 (2009) 1763–1772
http://dx.doi.org:/10.1016/j.bcp.2009.01.014

Pulmonary permeability edema is a major complication of acute lung injury (ALI), severe pneumonia and ARDS. This pathology can be accompanied by

(1) a reduction of alveolar liquid clearance capacity, caused by an inhibition of the expression of crucial sodium transporters, such as the epithelial sodium channel (ENaC) and the Na+-K+-ATPase,
(2) an epithelial and endothelial hyperpermeability and
(3) a disruption of the epithelial and endothelial barriers, caused by increased apoptosis or necrosis.

Since, apart from ventilation strategies, no standard treatment exists for permeability edema, the following chapters will review a selection of novel approaches aiming to improve these parameters in the capillary endothelium and the alveolar epithelium.

Apoptosis is an essential physiological process for the selective elimination of cells. However, the dysregulation of apoptotic pathways is thought to play an important role in the pathogenesis of ALI. Both delayed neutrophil apoptosis and enhanced endothelial/epithelial cell apoptosis have been identified in ALI/ARDS. In the case of neutrophils, which contribute significantly to ALI/ ARDS, studies in both animals and ARDS patients suggest that apoptosis is inhibited during the early stages (<2 h) of inflammation.

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors belonging to the nuclear hormone receptor superfamily, that includes receptors for steroid hormones, thyroid hormones, retinoic acid, and fat-soluble vitamins. Since their discovery in 1990, increasing data has been published on the role of PPARs in diverse processes, including lipid and glucose metabolism, diabetes and obesity, atherosclerosis, cellular proliferation and differentiation, neurological diseases, inflammation and immunity. PPARs have both gene-dependent and gene-independent effects. Gene-dependent functions involve the formation of heterodimers with the retinoid X-receptor. Activation by PPAR ligands results in the binding of the heterodimer to peroxisome proliferator response elements, located in the promoter regions of PPAR-regulated genes. Gene independent effects involve the direct binding of PPARs to transcription factors, such as NF-kB, which then alters their binding to DNA promoter elements. PPARs can also bind and sequester various cofactors for transcription factors, and thus further alter gene expression. Importantly, the precise effects of PPARs vary greatly between cell types. To date, three subtypes of PPAR have been identified: α, β, and ϒ. There is increasing data suggesting that PPAR signaling may play an important role in the pathobiology of systemic vascular disease. However, there is less data implicating PPAR signaling in diseases of the lung.

A role for PPARs in the control of inflammation was first evidenced for PPARα, where mice deficient in PPARα exhibited an increased duration of ear-swelling in response to the proinflammatory mediator, LTB4. More recently, a number of studies in mice and in humans have shown that PPAR agonists exhibit anti-inflammatory effects under a wide range of conditions. There are two main mechanisms by which PPARs exert their anti-inflammatory effect. The first involves complex formation, and the inhibition of transcription factors that positively regulate the transcription of pro-inflammatory genes. These include nuclear factor-kB (NF-kB), signal transducers and activators of transcription (STATs), nuclear factor of activated T cells (NF-AT), CAAT/enhancer binding protein (C/EBP) and activator protein 1 (AP-1). These transcription factors are the main mediators of the major proinflammatory cytokines, chemokines, and adhesion molecules involved in inflammation. The second PPAR-mediated anti-inflammatory pathway is mediated by the sequestration of rate limiting, but essential, co-activators or co-repressors.

Recent studies have shown that PPAR signaling can attenuate the airway inflammation induced by LPS in the mouse. It was shown that mice treated with the PPARα agonist, fenofibrate, had decreases in both inflammatory cell infiltration and inflammatory mediators. Conversely, PPARα -/- mice have been shown to have a greater number of neutrophils and macrophages, and increased levels of inflammatory mediators in bronchoalveolar lavage fluids (BALF). Other PPAR agonists, such as rosiglitazone or SB 21994 have also been shown to reduce LPS-mediated ALI in the mouse lung. PPARϒ signaling has also been shown to be protective in regulating pulmonary inflammation associated with fluorescein isothiocyanate (FITC)-induced lung injury, with the PPARϒ ligand pioglitazone decreasing neutrophil infiltration. Collectively, these data suggest that therapeutic agents that activate either or both PPARα and PPARϒ could be beneficial for the treatment of ALI.

Permeability edema is characterized by a reduced alveolar liquid clearance capacity, combined with an endothelial hyperpermeability. Various signaling pathways, such as those involving reactive oxygen species (ROS), Rho GTPases and tyrosine phosphorylation of junctional proteins, converge to regulate junctional permeability, either by affecting the stability of junctional proteins or by modulating their interactions. The regulation of junctional permeability is mainly mediated by dynamic interactions between the proteins of the adherens junctions and the actin cytoskeleton. Actin-mediated endothelial cell contraction is the result of myosin light chain (MLC) phosphorylation by MLC kinase (MLCK) in a Ca2+/calmodulin-dependent manner. RhoA additionally potentiates MLC phosphorylation, by inhibiting MLC phosphatase activity through its downstream effector Rho kinase (ROCK). As such, actin/myosin-driven contraction will generate a contractile force that pulls VE-cadherin inward. This contraction will force VE-cadherin to dissociate from its adjacent partner, as such producing interendothelial gaps.

Vascular endothelial cells can be regulated by nucleotides released from platelets. During vascular injury, broken cells are also the source of the extracellular nucleotides. Furthermore, endothelium may provide a local source of ATP within vascular beds. Primary cultures of human endothelial cells derived from multiple blood vessels release ATP constitutively and exclusively across the apical membrane under basal conditions. Hypotonic challenge or the calcium agonists (ionomycin and thapsigargin) stimulate ATP release in a reversible and regulated manner. Enhanced release of pharmacologically relevant amounts of ATP was observed in endothelial cells under such stimuli as shear stress, lipopolysaccharide (LPS), and ATP itself. Pearson and Gordon demonstrated that incubation of aortic endothelial and smooth muscle cells with thrombin resulted in the specific release of ATP, which was converted to ADP by vascular hydrolases. Yang et al. showed that endothelial cells isolated from guinea pig heart release nucleotides in response to bradykinin, acetylcholine, serotonin and ADP. Nucleotide action is mediated by cell surface purinoreceptors. Once released from endothelial cells, ATP may act in the blood vessel lumen at P2 receptors on nearby endothelium downstream from the site of release. ATP is also degraded rapidly and its metabolites have also been recognized as signaling molecules, which can initiate additional receptor-mediated functions. These include ADP and the final hydrolysis product adenosine.

Signal transduction pathways implicated in ATP-mediated endothelial barrier enhancement

Signal transduction pathways implicated in ATP-mediated endothelial barrier enhancement

Signal transduction pathways implicated in ATP-mediated endothelial barrier enhancement

During the course of ALI, the alveolar space, as well as the interstitium, are sites of intense inflammation, leading to the local production of pro-inflammatory cytokines, such as IL-1β, TGF-β and TNF. The latter pleiotropic cytokine is a 51 kDa homotrimeric protein, binding to two types of receptors, i.e. TNF-R1 and TNF-R2 and which is mainly produced by activated macrophages and T cells. Soluble TNF, as well as the soluble TNF receptors 1 and 2, are generated upon cleavage of membrane TNF or of the membrane associated receptors, respectively, by the enzyme TNF-α convertase (TACE). TNF-R1, but not TNF-R2, contains a death domain, which signals apoptosis upon the formation of the Death Inducing Signaling Complex (DISC). In spite of its lack of a death domain, TNF-R2 can nevertheless be implicated in apoptosis induction, since its activation causes degradation of TNF Receptor Associated Factor 2 (TRAF2), an inhibitor of the TNF-R1-induced DISC formation. Moreover, apoptosis induction of lung microvascular endothelial cells by TNF was shown to require activation of both TNF receptors. TNF-R2 was also shown to be important for ICAM-1 upregulation in endothelial cells in vitro and in vivo, an activity important in the sequestration of leukocytes in the microvessels. Moreover, lung microvascular endothelial cells isolated from ARDS patients express significantly higher levels of TNF-R2 and of ICAM-1 than cells isolated from patients who had undergone a lobectomy for lung carcinoma, used as controls. These findings therefore suggest that ICAM-1 and TNF-R2 may have a particular involvement in the pathogenesis of acute lung injury.

Dichotomous activity of TNF in alveolar liquid clearance and barrier protection

Dichotomous activity of TNF in alveolar liquid clearance and barrier protection

Dichotomous activity of TNF in alveolar liquid clearance and barrier protection during ALI. TNF, which is induced during ALI, causes a downregulation of ENaC expression in type II alveolar epithelial cells, upon activating TNF-R1. Moreover, TNF increases permeability, by means of interfering with tight junctions (TJ) in both alveolar epithelial (AEC) and capillary endothelial cells (MVEC). ROS, the generation of which is frequently increased during ALI, were also shown to downregulate ENaC and Na+-K+-ATPase expression and moreover also lead to decreased endothelial barrier integrity. The TIP peptide, mimicking the lectin-like domain of TNF, is able to increase sodium uptake in alveolar epithelial cells and to restore endothelial barrier integrity, as such providing a significant protection against the development of permeability edema (red lines: inhibition, green arrows: activation).

Proposed mechanism of action for the anti-inflammatory and barrier-protective actions of hsp90 inhibitors.

Proposed mechanism of action for the anti-inflammatory and barrier-protective actions of hsp90 inhibitors.

Proposed mechanism of action for the anti-inflammatory and barrier-protective actions of hsp90 inhibitors.

Permeability edema represents a life-threatening complication of acute lung injury, severe pneumonia and ARDS, characterized by a combined dysregulation of pulmonary epithelial and endothelial apoptosis, endothelial barrier integrity and alveolar liquid clearance capacity. As such, it is likely that several of these parameters have to be targeted in order to obtain a successful therapy. This review focuses on a selection of recently discovered substances and mechanisms that might improve ALI therapy. As such, we have discussed the inhibition of apoptosis and necrosis occurring during ALI, by means of the restoration of Zn2+ homeostasis. PPARα and ϒ agonists can represent therapeutically  promising molecules, since they inhibit transcription factors as well as essential co-activators involved in the activation of pro-inflammatory cytokines, chemokines and adhesion molecules, all of which are implicated in ALI. Apart from inducing a potent inhibition of inflammation upon interfering with NF-kB activation, hsp90 inhibitors were shown to prevent and restore endothelial barrier integrity. These agents are able to significantly improve survival and lung function during LPS-induced ALI. A restoration of endothelial barrier integrity during ALI can also be obtained upon increasing extracellular levels of ATP or adenosine, which activate the purinoreceptors P2Y and P1A2, respectively, leading to a decrease in myosin light chain phosphorylation and an increase in MLC phosphatase 1 activity. The pro-inflammatory cytokine TNF is involved in endothelial apoptosis and hyperpermeability, as well as in the reduction of alveolar liquid clearance, upon activating its receptors. However, apart from its receptor binding sites, TNF harbors a lectin-like domain, which can be mimicked by the TIP peptide. This peptide has been shown to increase alveolar liquid clearance and moreover induces endothelial barrier protection. As such, TNF can be considered as a moonlighting cytokine, combining both positive and negative activities for permeability edema generation within one molecule.

 

The protective effect of CDDO-Me on lipopolysaccharide-induced acute lung injury in mice

Tong Chen, Yi Moua, Jiani Tan, LinlinWei, Yixue Qiao, Tingting Wei, et al.
International Immunopharmacology 25 (2015) 55–64
http://dx.doi.org/10.1016/j.intimp.2015.01.011

ALI is a clinical syndrome characterized by a disruption of epithelial integrity, neutrophil accumulation, noncardiogenic pulmonary edema, severe hypoxemia and an intense pulmonary inflammatory response with a wide array of increasing severity of lung parenchymal injury. Previous studies have shown that lots of pathogenesis contribute to ALI, such as oxidant/antioxidant dysfunction, dysregulation of inflammatory/anti-inflammatory pathway, upregulation of chemokine production and adhesion molecules. However, to date there is no effective medicine to control ALI. Lipopolysaccharide (LPS) is a main component of the outer membrane of Gram negative bacteria. It has been reported to activate toll like receptors 4 (TLR4) and to stimulate the release of inflammatory mediators inducing ALI-like symptoms. Intratracheal administration of LPS has been used to construct animal models of ALI.

The biological importance of naturally occurring triterpenoids has long been recognized. Oleanolic acid, exhibiting modest biological activities, has been marketed in China as an oral drug for the treatment of liver disorders in humans. Among its derivatives, bardoxolonemethyl (2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid methylester) CDDO-Me, had completed a successful phase I clinical trial for the treatment of cancer and started a phase II trial for the treatment of patients with pulmonary arterial hypertension. For its broad spectrum antiproliferative and anti-tumorigenic activities, CDDO-Me has also been reported to possess a number of pharmacological activities such as antioxidant, anti-tumor and anti-inflammatory effects. However, the mechanisms by which CDDO-Me exerted its anti-inflammatory effects on macrophage were insufficiently elucidated. More importantly, there is no available report to evaluate its therapeutic effect on acute lung injury.

CDDO-Me, initiated in a phase II clinical trial, is a potential useful therapeutic agent for cancer and inflammatory dysfunctions, whereas the therapeutic efficacy of CDDO-Me on LPS-induced acute lung injury (ALI) has not been reported as yet. The purpose of the present study was to explore the protective effect of CDDO-Me on LPS-induced ALI in mice and to investigate its possible mechanism. BalB/c mice received CDDO-Me (0.5 mg/kg, 2 mg/kg) or dexamethasone (5 mg/kg) intraperitoneally 1 h before LPS stimulation and were sacrificed 6 h later. W/D ratio, lung MPO activity, number of total cells and neutrophils, pulmonary histopathology, IL-6, IL-1β, and TNF-α in the BALF were assessed. Furthermore, we estimated iNOS, IL-6, IL-1β, and TNF-α mRNA expression and NO production as well as the activation of the three main MAPKs, AkT, IκB-α and p65. Pretreatment with CDDO-Me significantly ameliorated W/D ratio, lung MPO activity, inflammatory cell infiltration, and inflammatory cytokine production in BALF from the in vivo study. Additionally, CDDO-Me had beneficial effects on the intervention for pathogenesis process at molecular, protein and transcriptional levels in vitro. These analytical results provided evidence that CDDO-Me could be a potential therapeutic candidate for treating LPS-induced ALI.

Effects of CDDO-Me on LPS-mediated lung changes

Effects of CDDO-Me on LPS-mediated lung histopathologic changes in lung tissues. (A) The lung section from the control mice; (B) the lung section from the mice administered with LPS (8 mg/kg); (C) the lung section from the mice administered with dexamethasone (5 mg/kg) and LPS (8 mg/kg); (D) the lung section from the mice administered with CDDO-Me (0.5mg/kg) and LPS (8mg/kg); (E) the lung section from the mice administered with CDDO-Me (2mg/kg) and LPS (8mg/kg); (hematoxylin and eosin staining, magnification 200×). Control group: the green arrow indicated alveolar wall, no hyperemia. All the other groups: The black arrow indicated the inflammatory cell infiltration; the green arrow indicated alveolar wall hyperemia.

 

The impact of cardiac dysfunction on acute respiratory distress syndrome and mortality in mechanically ventilated patients with severe sepsis and septic shock: An observational study

Brian M. Fuller, Nicholas M. Mohr, Thomas J. Graetz, et al.
Journal of Critical Care 30 (2015) 65–70
http://dx.doi.org/10.1016/j.jcrc.2014.07.027

Purpose: Acute respiratory distress syndrome (ARDS) is associated with significant mortality and morbidity in survivors. Treatment is only supportive, therefore elucidating modifiable factors that could prevent ARDS could have a profound impact on outcome. The impact that sepsis-associated cardiac dysfunction has on ARDS is not known. Materials and Methods: In this retrospective observational cohort study of mechanically ventilated patients with severe sepsis and septic shock, 122 patients were assessed for the impact of sepsis-associated cardiac dysfunction on incidence of ARDS (primary outcome) and mortality. Results: Sepsis-associated cardiac dysfunction occurred in 44 patients (36.1%). There was no association of sepsis-associated cardiac dysfunction with ARDS incidence (p= 0.59) or mortality, and no association with outcomes in patients that did progress to ARDS after admission. Multivariable logistic regression demonstrated that higher BMI was associated with progression to ARDS (adjusted OR 11.84, 95% CI 1.24 to 113.0, p= 0.02). Conclusions: Cardiac dysfunction in mechanically ventilated patients with sepsis did not impact ARDS incidence, clinical outcome in ARDS patients, or mortality. This contrasts against previous investigations demonstrating an influence of nonpulmonary organ dysfunction on outcome in ARDS. Given the frequency of ARDS as a sequela of sepsis, the impact of cardiac dysfunction on outcome should be further studied.

 

Suppression of NF-κβ pathway by crocetin contributes to attenuation of lipopolysaccharide-induced acute lung injury in mice

Ruhui Yang, Lina Yang, Xiangchun Shen, Wenyuan Cheng, et al.
European Journal of Pharmacology 674 (2012) 391–396
http://dx.doi.org:/10.1016/j.ejphar.2011.08.029

Crocetin, a carotenoid compound, has been shown to reduce expression of inflammation and inhibit the production of reactive oxygen species. In the present study, the effect of crocetin on acute lung injury induced by lipopolysaccharide (LPS) was investigated in vivo. In the mouse model, pretreatment with crocetin at dosages of 50 and 100 mg/kg reduced the LPS-induced lung edema and histological changes, increased LPS-impaired superoxide dismutase (SOD) activity, and decreased lung myeloperoxidase (MPO) activity. Furthermore, treatment with crocetin significantly attenuated LPS-induced mRNA and the protein expressions of interleukin-6 (IL-6), macrophage chemoattractant protein-1 (MCP-1), and tumour necrosis factor-α (TNF-α) in lung tissue. In addition, crocetin at different dosages reduced phospho-IκB expression and NF-κB activity in LPS-induced lung tissue alteration. These results indicate that crocetin can provide protection against LPS-induced acute lung injury in mice.

 

Sauchinone, a lignan from Saururus chinensis, attenuates neutrophil pro-inflammatory activity and acute lung injury

Hui-Jing Han, Mei Li, Jong-Keun Son, Chang-Seob Seo, et al.
International Immunopharmacology 17 (2013) 471–477
http://dx.doi.org/10.1016/j.intimp.2013.07.011

Previous studies have shown that sauchinone modulates the expression of inflammatory mediators through mitogen-activated protein kinase (MAPK) pathways in various cell types. However, little information exists about the effect of sauchinone on neutrophils, which play a crucial role in inflammatory process such as acute lung injury (ALI). We found that sauchinone decreased the phosphorylation of p38 MAPK in lipopolysaccharide (LPS)-stimulated murine bone marrow neutrophils, but not ERK1/2 and JNK. Exposure of LPS-stimulated neutrophils to sauchinone or SB203580, a p38 inhibitor, diminished production of tumor necrosis factor (TNF)-α and macrophage inflammatory protein (MIP)-2 compared to neutrophils cultured with LPS. Treatment with sauchinone decreased the level of phosphorylated ribosomal protein S6 (rpS6) in LPS-stimulated neutrophils. Systemic administration of sauchinone to mice led to reduced levels of phosphorylation of p38 and rpS6 in mice lungs given LPS, decreased TNF-α and MIP-2 production in bronchoalveolar lavage fluid, and also diminished the severity of LPS-induced lung injury, as determined by reduced neutrophil accumulation in the lungs, wet/dry weight ratio, and histological analysis. These results suggest that sauchinone diminishes LPS-induced neutrophil activation and ALI.

In the present study, the systemic administration of sauchinone decreased the phosphorylation of p38 MAPK and rpS6 in mice lungs subjected to LPS and diminished the severity of LPS-induced ALI. Neutrophils play an important role in acute inflammatory processes, such as ALI, which was demonstrated by various experimental models. Previous reports suggested that p38 MAPK inhibition of murine neutrophils could lead to the loss of chemotaxis toward MIP-2, as well as the loss of TNF-αandMIP-2 production in response to LPS, and also attenuated neutrophil accumulation in LPS-induced ALI models. Therefore, the beneficial effects of sauchinone on LPS-induced ALI are likely associated with decreases in the production of pro-inflammatory mediators by neutrophils, consistent with our in vitro experiments. However, we cannot exclude that the effects of sauchinone on reducing the release of TNF-α and MIP-2 in mice lungs subjected to LPS, with the resultant prevention of ALI, could be affected by various pulmonary cell populations, such as alveolar macrophages. Also, the inhibitory effects of sauchinone on NF-κB activation through various pulmonary cell populations (Supplemental Fig. S2), in addition to p38MAPK activity in mouse lungs given LPS, might enhance the anti-inflammatory action of sauchinone in mouse lungs subjected to LPS. In conclusion, we found that sauchinone significantly diminished the release of inflammatory mediators in isolated neutrophils and lungs subjected to LPS. The anti-inflammatory action of sauchinone was associated with the prevention of p38 MAPK and rpS6 activation. These findings suggest that sauchinone may be an appropriate pharmacological candidate for the treatment of ALI as well as other neutrophil driven acute inflammatory diseases.
Supplementary data to this article can be found online at
http://dx.doi.org/10.1016/j.intimp.2013.07.011

 

Protective effect of dexmedetomidine in a rat model of α-naphthylthiourea- induced acute lung injury

Volkan Hancı, Gamze Yurdakan, Serhan Yurtlu, et al.
J Surg Res 178 (2012):424-430
http://dx.doi.org:/10.1016/j.jss.2012.02.027

Background: We assessed the effects of dexmedetomidine in a rat model of a-naphthylthiourea (ANTU)einduced acute lung injury.  Methods: Forty Wistar Albino male rats weighing 200e240 g were divided into 5 groups (n = 8 each), including a control group. Thus, there were one ANTU group and three dexmedetomidine groups (10-, 50-, and 100-mg/kg treatment groups), plus a control group. The control group provided the normal base values. The rats in the ANTU group were given 10 mg/kg of ANTU intraperitoneally and the three treatment groups received 10, 50, or 100 mg/kg of dexmedetomidine intraperitoneally 30 min before ANTU application. The rat body weight (BW), pleural effusion (PE), and lung weight (LW) of each group were measured 4 h after ANTU administration. The histopathologic changes were evaluated using hematoxylin-eosin staining. Results: The mean PE, LW, LW/BW, and PE/BW measurements in the ANTU group were significantly greater than in the control groups and all dexmedeto-midine treatment groups (P < 0.05). There were also significant decreases in the mean PE, LW, LW/BW and PE/BW values in the dexmedetomidine 50-mg/kg group compared with those in the ANTU group (P < 0.01). The inflammation, hemorrhage, and edema scores in the ANTU group were significantly greater than those in the control or dexmedetomidine 50-mg/kg group (P < 0.01). Conclusion: Dexmedetomidine treatment has demonstrated  a potential benefit by preventing ANTU-induced acute lung injury in an experimental rat model. Dexmedetomidine could have a potential protective effect on acute lung injury in intensive care patients.

 

Protective effects of Isofraxidin against lipopolysaccharide-induced acute lung injury in mice

Xiaofeng Niu, YuWang, Weifeng Li, Qingli Mu, et al.
International Immunopharmacology 24 (2015) 432–439
http://dx.doi.org/10.1016/j.intimp.2014.12.041

Acute lung injury (ALI) is a life-threatening disease characterized by serious lung inflammation and increased capillary permeability, which presents a high mortality worldwide. Isofraxidin (IF), a Coumarin compound isolated from the natural medicinal plants such as Sarcandra glabra and Acanthopanax senticosus, has been reported to have definite anti-bacterial, anti-oxidant, and anti-inflammatory activities. However, the effects of IF against lipopoly-saccharide-induced ALI have not been clarified. The aim of the present study is to explore the protective effects and potential mechanism of IF against LPS-induced ALI in mice. In this study, We found that pretreatment with IF significantly lowered LPS-induced mortality and lung wet-to-dry weight (W/D) ratio and reduced the levels of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and prostaglandin E2 (PGE2) in serum and bronchoalveolar lavage fluid (BALF). We also found that total cells, neutrophils and macrophages in BALF,MPO activity in lung tissues were markedly decreased. Besides, IF obviously inhibited lung histopathological changes and cyclooxygenase-2 (COX-2) protein expression. These results suggest that IF has a protective effect against LPS induced ALI, and the protective effect of IF seems to result from the inhibition of COX-2 protein expression in the lung, which regulates the production of PGE2.

Ingestion of LPS stimulates vascular permeability, promotes inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) from blood into lung tissues and activates numerous inflammatory cells such as neutrophils and macrophages. In macrophages, LPS challenge induces the transcription of gene encoding pro-inflammatory protein, which leads to cytokine release and synthesis of enzymes, such as cyclo-oxygenase-2 (COX-2). COX-2 usually can’t be found in normal tissues, but widely induced by pro-inflammatory stimuli, such as cytokines, endotoxins, and growth factors. COX-2 plays a vital role in the regulation of inflammatory process by modulating the production of prostaglandin E2 (PGE2). PGE2, induced by cytokines and other initiator, is an inflammatory mediator which is produced in the regulation of COX-2. Previous researches demonstrated that inhibition of COX-2 produced a dramatically anti-inflammatory effect with little gastrointestinal toxicity. Therefore, inhibition of COX-2 protein expression has far-reaching significance in the treatment of ALI.

effects of IF on LPS-induced mortality in ALI mice

effects of IF on LPS-induced mortality in ALI mice

The effects of IF on LPS-induced mortality in ALI mice (n = 12/group). IF (5, 10, 15 mg/kg, i.p.) or DEX (5 mg/kg, i.p.) were given to mice 1 h prior to LPS challenge. The mortalities were observed at 0, 12, 24, 36, 48, 60, and 72 h. ###P = 0.001 when compared with the control group; *P = 0.05, **P = 0.01, and ***P = 0.001 when compared with the LPS group.

 

Protective effects of intranasal curcumin on paraquot induced acute lung injury (ALI) in mice

Namitosh Tyagi, Asha Kumaria, D. Dash, Rashmi Singh
Environment  Toxicol  & Pharmacol  38 (2014) 913–921
http://dx.doi.org/10.1016/j.etap.2014.10.003

Paraquot (PQ) is widely and commonly used as herbicide and has been reported to be hazardous as it causes lung injury. However, molecular mechanism underlying lung toxicity caused by PQ has not been elucidated. Curcumin, a known anti-inflammatory molecule derived from rhizomes of Curcuma longa has variety of pharmacological activities including free-radical scavenging properties but the protective effects of curcumin on PQ-induced acute lung injury (ALI) have not been studied. In this study, we aimed to study the effects of curcumin on ALI caused by PQ in male parke’s strain mice which were challenged acutely byPQ (50 mg/kg, i.p.) with or without curcumin an hour before (5 mg/kg, i.n.) PQ intoxication. Lung specimens and the bronchoalveolar lavage fluid (BALF) were isolated for pathological and biochemical analysis after 48 h of PQ exposure. Curcumin administration has significantly enhanced superoxide dismutase (SOD) and catalase activities. Lung wet/dry weight ratio, malondialdehyde (MDA) and lactate dehydrogenase (LDH) content, total cell number and myeloperoxidase (MPO) levels in BALF as well as neutrophil infiltration were attenuated by curcumin. Pathological studies also revealed that intranasal curcumin alleviate PQ-induced pulmonary damage and pro-inflammatory cytokine levels like tumor necrosis factor-α (TNF-α) and nitric oxide (NO). These results suggest that intranasal curcumin may directly target lungs and curcumin inhalers may prove to be effective in PQ-induced ALI treatment in near future.

 

Phillyrin attenuates LPS-induced pulmonary inflammation via suppression of MAPK and NF-κB activation in acute lung injury mice

Wei-ting Zhong, Yi-chun Wu, Xian-xing Xie, Xuan Zhou, et al.
Fitoterapia 90 (2013) 132–139
http://dx.doi.org/10.1016/j.fitote.2013.06.003

Phillyrin (Phil) is one of the main chemical constituents of Forsythia suspensa (Thunb.), which has shown to be an important traditional Chinese medicine. We tested the hypothesis that Phil modulates pulmonary inflammation in an ALI model induced by LPS. Male BALB/c mice were pretreated with or without Phil before respiratory administration with LPS, and pretreated with dexamethasone as a control. Cytokine release (TNF-α, IL-1β, and IL-6) and amounts of inflammatory cell in bronchoalveolar lavage fluid (BALF) were detected by ELISA and cell counting separately. Pathologic changes, including neutrophil infiltration, interstitial edema, hemorrhage, hyaline membrane formation, necrosis, and congestion during acute lung injury in mice were evaluated via pathological section with HE staining. To further investigate the mechanism of Phil anti-inflammatory effects, activation of MAPK and NF-κB pathways was tested by western blot assay. Phil pretreatment significantly attenuated LPS-induced pulmonary histopathologic changes, alveolar hemorrhage, and neutrophil infiltration. The lung wet-to-dry weight ratios, as the index of pulmonary edema, were markedly decreased by Phil retreatment. In addition, Phil decreased the production of the proinflammatory cytokines including (TNF-α, IL-1β, and IL-6) and the concentration of myeloperoxidase (MPO) in lung tissues. Phil pretreatment also significantly suppressed LPS-induced activation of MAPK and NF-κB pathways in lung tissues. Taken together, the results suggest that Phil may have a protective effect on LPS-induced ALI, and it potentially contributes to the suppression of the activation of MAPK and NF-κB pathways. Phil may be a new preventive agent of ALI in the clinical setting.

A mass of studies have been reported basically on alleviating LPS-induced acute lung injury in models. Phillyrin (Fig. 1), a lignin, is one of the main chemical constituents of Forsythia suspensa (Thunb.), which is an important traditional Chinese medicine (“Lianqiao” in Chinese), and has long been used for gonorrhea, erysipelas, inflammation, pyrexia and ulcer. Previous studies indicated that Phil significantly inhibited NO production in LPS-activated macrophage cells. But there is not much evidence showing the anti-inflammatory properties of phillyrin. In the present study, we sought to investigate the effects of phillyrin on LPS-induced pulmonary inflammation in mice.

Fig. not shown. A: Effects of Phil on histopathological changes in lung tissues in LPS-induced ALI mice. Mice were given an intragastric administration of Phil (10 and 20 mg/kg) or Dex (5 mg/kg) 1 h prior to an intranasal administration of LPS. Then mice were anesthetized and lung tissue samples were collected at 6 h after LPS challenge for histological evaluation. These representative histological changes of the lung were obtained from mice of different groups (hematoxylin and eosin staining, original magnification 200×, Scale bar: 50 μm). B: Effects of Phil on LPS-induced lung morphology. The slides were histopathologically evaluated using a semi-quantitative scoring method. Lung injury was graded from 0 (normal) to 4 (severe) in four categories: congestion, edema, interstitial inflammation and inflammatory cell infiltration. The total lung injury score was calculated by adding up the individual scores of each category. The values presented are the means ± S.E.M. (n = 4–6 in each group). ##P b 0.01 vs. the control group, **P b 0.01 vs. the LPS group. Cont: control group; LPS: LPS group; Phil + LPS: Phil + LPS group; Dex + LPS: Dex + LPS group.

In summary, the present study indicated that Phil has a protective effect on LPS-induced acute lung injury. Phil significantly attenuated histopathological changes initiated by LPS via reducing over inflammatory responses. We also demonstrated that MAPK and NF-κB signaling pathways are the important targets of Phil to perform its actions. Phil acts by preventing NF-κB translocation to the nucleus or inhibiting the activation of MAPKs directly or indirectly, which is to be investigated in further studies. All these results suggest that Phil may be a new therapeutic agent for the prevention of inflammation during acute lung injury.

 

 

 

 

Read Full Post »

The Reconstruction of Life Processes requires both Genomics and Metabolomics to explain Phenotypes and Phylogenetics

Writer and Curator: Larry H. Bernstein, MD, FCAP 

 

phylogenetics

phylogenetics

http://upload.wikimedia.org/wikipedia/commons/thumb/1/12/CollapsedtreeLabels-simplified.svg/200px-CollapsedtreeLabels-simplified.svg.png

 

This discussion that completes and is an epicrisis (summary and critical evaluation) of the series of discussions that preceded it.

  1. Innervation of Heart and Heart Rate
  2. Action of hormones on the circulation
  3. Allogeneic Transfusion Reactions
  4. Graft-versus Host reaction
  5. Unique problems of perinatal period
  6. High altitude sickness
  7. Deep water adaptation
  8. Heart-Lung-and Kidney
  9. Acute Lung Injury

The concept inherent in this series is that the genetic code is an imprint that is translated into a message.  It is much the same as a blueprint, or a darkroom photographic image that has to be converted to a print. It is biologically an innovation of evolutionary nature because it establishes a simple and reproducible standard for the transcription of the message through the transcription of the message using strings of nucleotides (oligonucleotides) that systematically transfer the message through ribonucleotides that communicate in the cytoplasm with the cytoskeleton based endoplasmic reticulum (ER), composing a primary amino acid sequence.  This process is a quite simple and convenient method of biological activity.  However, the simplicity ends at this step.  The metabolic components of the cell are organelles consisting of lipoprotein membranes and a cytosol which have particularly aligned active proteins, as in the inner membrane of the mitochondrion, or as in the liposome or phagosome, or the structure of the  ER, each of which is critical for energy transduction and respiration, in particular, for the mitochondria, cellular remodeling or cell death, with respect to the phagosome, and construction of proteins with respect to the ER, and anaerobic glycolysis and the hexose monophosphate shunt in the cytoplasmic domain.  All of this refers to structure and function, not to leave out the membrane assigned transport of inorganic, and organic ions (electrolytes and metabolites).

I have identified a specific role of the ER, the organelles, and cellular transactions within and between cells that is orchestrated.  But what I have outlined is a somewhat limited and rigid model that does not reach into the dynamics of cellular transactions.  The DNA has expression that may be old, no longer used messages, and this is perhaps only part of a significant portion of “dark matter”.  There is also nuclear DNA that is enmeshed with protein, mRNA that is a copy of DNA, and mDNA  is copied to ribosomal RNA (rRNA).  There is also rDNA. The classic model is DNA to RNA to protein.  However, there is also noncoding RNA, which plays an important role in regulation of transcription.

This has been discussed in other articles.  But the important point is that proteins have secondary structure through disulfide bonds, which is determined by position of sulfur amino acids, and by van der Waal forces, attraction and repulsion. They have tertiary structure, which is critical for 3-D structure.  When like subunits associate, or dissimilar oligomers, then you have heterodimers and oligomers.  These constructs that have emerged over time interact with metabolites within the cell, and also have an important interaction with the extracellular environment.

When you take this into consideration then a more complete picture emerges. The primitive cell or the multicellular organism lives in an environment that has the following characteristics – air composition, water and salinity, natural habitat, temperature, exposure to radiation, availability of nutrients, and exposure to chemical toxins or to predators.  In addition, there is a time dimension that proceeds from embryonic stage to birth in mammals, a rapid growth phase, a tapering, and a decline.  The time span is determined by body size, fluidity of adaptation, and environmental factors.  This is covered in great detail in this work.  The last two pieces are in the writing stage that completes the series. Much content has already be presented in previous articles.

The function of the heart, kidneys and metabolism of stressful conditions have already been extensively covered in http://pharmaceuticalintelligence.com  in the following and more:

The Amazing Structure and Adaptive Functioning of the Kidneys: Nitric Oxide – Part I

http://pharmaceuticalintelligence.com/2012/11/26/the-amazing-structure-and-adaptive-functioning-of-the-kidneys/

Nitric Oxide and iNOS have Key Roles in Kidney Diseases – Part II

http://pharmaceuticalintelligence.com/2012/11/26/nitric-oxide-and-inos-have-key-roles-in-kidney-diseases/

The pathological role of IL-18Rα in renal ischemia/reperfusion injury – Nature.com

http://pharmaceuticalintelligence.com/2014/10/24/the-pathological-role-of-il-18r%CE%B1-in-renal-ischemiareperfusion-injury-nature-com/

Summary, Metabolic Pathways

http://pharmaceuticalintelligence.com/2014/10/23/summary-metabolic-pathways/

 

Read Full Post »

Altitude Adaptation

Writer and Curator: Larry H. Bernstein, MD, FCAP 

 

Introduction

Land adapted animals depend on respiration for oxygen supply, but have adapted to altitudes that have difference oxygen contents.  In this discussion we explore how animals have adapted to oxygen supply in different terrestrial habitats, and also how humans adjust to short term changes in high and extreme altitudes.

High-altitude adaptation is an evolutionary modification in animals, most notably in birds and mammals, by which species are subjected to considerable physiological changes to survive in extremely high mountainous environments. As opposed to short-term adaptation, or more properly acclimatization (which is basically an immediate physiological response to changing environment), the term “high-altitude adaptation” has strictly developed into the description of an irreversible, long-term physiological responses to high-altitude environments, associated with heritable behavioral and genetic changes. Perhaps, the phenomenon is most conspicuous, at least best documented, in human populations such as the Tibetans, the South Americans and the Ethiopians, who live in the otherwise uninhabitable high mountains of the Himalayas, Andes and Ethiopia respectively; and this represents one of the finest examples of natural selection in action.

Oxygen, essential for animal life, is proportionally abundant in the atmosphere with height from the sea level; hence, the highest mountain ranges of the world are considered unsuitable for habitation. Surprisingly, some 140 million people live permanently at high altitudes (>2,500 m) in North, Central and South America, East Africa, and Asia, and flourish very well for millennia in the exceptionally high mountains, without any apparent complications. This has become a recognized instance of the process of Darwinian evolution in humans acting on favorable characters such as enhanced respiratory mechanisms. As a matter of fact, this adaptation is so far the fastest case of evolution in humans that is scientifically documented. Among animals only few mammals (such as yak, ibex, Tibetan gazelle, vicunas, llamas, mountain goats, etc.) and certain birds are known to have completely adapted to high-altitude environments.

These adaptations are an example of convergent evolution, with adaptations occurring simultaneously on three continents. Tibetan humans and Tibetan domestic dogs found the genetic mutation in both species, EPAS1. This mutation has not been seen in Andean humans, showing the effect of a shared environment on evolution.

At elevation higher than 8,000 metres (26,000 ft), which is called the “death zone” in mountaineering, the available oxygen in the air is so low that it is considered insufficient to support life. And higher than 7,600 m is seriously lethal. Yet, there are Tibetans, Ethiopians and Americans who habitually live at places higher than 2,500 m from the sea level. For normal human population, even a brief stay at these places means mountain sickness, which is a syndrome of hypoxia or severe lack of oxygen, with complications such as fatigue, dizziness, breathlessness, headaches, insomnia, malaise, nausea, vomiting, body pain, loss of appetite, ear-ringing, blistering and purpling and of the hands and feet, and dilated veins. Amazingly for the native highlanders, there are no adverse effects; in fact, they are perfectly normal in all respects. Basically, the physiological and genetic adaptations in these people involve massive modification in the oxygen transport system of the blood, especially molecular changes in the structure and functions hemoglobin, a protein for carrying oxygen in the body. This is to compensate for perpetual low oxygen environment. This adaptation is associated with better developmental patterns such as high birth weight, increased lung volumes, increased breathing, and higher resting metabolism.

http://en.wikipedia.org/wiki/High-altitude_adaptation

Acute Mountain Sickness: Pathophysiology, Prevention, and Treatment

Chris Imraya, Alex Wright, Andrew Subudhie,, Robert Roache
Progress in Cardiovascular Diseases 52 (2010) 467–484
http://dx.doi.org:/10.1016/j.pcad.2010.02.003

Barometric pressure falls with increasing altitude and consequently there is a reduction in the partial pressure of oxygen resulting in a hypoxic challenge to any individual ascending to altitude. A spectrum of high altitude illnesses can occur when the hypoxic stress outstrips the subject’s ability to acclimatize. Acute altitude-related problems consist of the common syndrome of acute mountain sickness, which is relatively benign and usually self-limiting, and the rarer, more serious syndromes of high-altitude cerebral edema and high-altitude pulmonary edema. A common feature of acute altitude illness is rapid ascent by otherwise fit individuals to altitudes above 3000 m without sufficient time to acclimatize. The susceptibility of an individual to high altitude syndromes is variable but generally reproducible. Prevention of altitude-related illness by slow ascent is the best approach, but this is not always practical. The immediate management of serious illness requires oxygen (if available) and descent of more than 300 m as soon as possible. In this article, we describe the setting and clinical features of acute mountain sickness and high altitude cerebral edema, including an overview of the known pathophysiology, and explain contemporary practices for both prevention and treatment exploring the comprehensive evidence base for the various interventions.

Acute mountain sickness (AMS) and high-altitude cerebral edema (HACE) strike people who travel too fast to high altitudes that lie beyond their current level of acclimatization. Understanding AMS and HACE is important because AMS can sharply limit recreation and work at high altitude. The syndromes can be identified early and reliably without sophisticated instruments, and when AMS and HACE are recognized early, most cases respond rapidly with complete recovery in a few hours (AMS) to days (HACE).

High-altitude headache (HAH) is the primary symptom of AMS. High-altitude headache in AMS usually occurs with some combination of other symptoms.
These are –  insomnia, fatigue (beyond that expected from the day’s activities), dizziness, anorexia, and nausea. The headache often worsens during the night and with exertion. Insomnia is the next most frequent complaint. Poor sleep can occur secondary to periodic breathing, severe headache, dizziness, and shortness of breath, among other causes. Anorexia and nausea are common, with vomiting reported less frequently in trekkers to 4243 m.

AMS is distinguished only by symptoms. The progression of AMS to HACE is marked by altered mental status, including impaired mental capacity, drowsiness, stupor, and ataxia. Coma may develop as soon as 24 hours after the onset of ataxia or change in mental status. The severity of AMS can be scored using the Lake Louise Questionnaire, or the more detailed Environmental Symptoms Questionnaire, or by the use of a simple analogue scale. Today, more than 100 years after the first clear clinical descriptions of AMS and HACE, we have advanced our understanding of the physiology of acclimatization to high altitude, and the pathophysiology of AMS and HACE.

As altitude increases, barometric pressure falls (see Fig ). This fall in barometric pressure causes a corresponding drop in the partial pressure of oxygen (21% of barometric pressure) resulting in hypobaric hypoxia. Hypoxia is the major challenge humans face at high altitude, and the primary cause of AMS and HACE. It follows that oxygen partial pressure is more important than
geographic altitude, as exemplified near the poles where the atmosphere is thinner and, thus, barometric pressure is lower. Lower barometric pressure at the poles can result in oxygen partial pressures that are physiologically equivalent to altitudes 100 to 200 m higher at more moderate latitudes. We define altitude regions as high altitude (1500-3500 m), very high altitude (3500-5500 m), and extreme altitude (>5500 m).

Neurological consequences of increasing altitude

Neurological consequences of increasing altitude

Neurological consequences of increasing altitude: The relation among altitude (classified as high [1500–3500 m], very high [3500-5500 m] and extreme [>5500 m]), the partial pressure of oxygen, and the neurological consequences of acute and gradual exposure to these pressure changes. Neurological consequences will vary greatly from person to person and with rate of ascent. HACE is far more common at higher altitudes, although there are case reports of HACE at 2500 m.

It is important for any discussion of AMS and HACE to have as a starting point an understanding of acclimatization. The process of acclimatization involves a series of adjustments by the body to meet the challenge of hypoxemia. While we have a general understanding of systemic changes associated with acclimatization, the underlying molecular and cellular processes are not yet fully described. Recent findings suggest that the process may be initiated by widespread molecular up-regulation of hypoxia inducible factor-1. Downstream processes ultimately act to offset hypoxemia, including elevated ventilation leading to a rise in arterial oxygen saturation (SaO2), a mild diuresis and contraction of plasma volume such that more oxygen is carried per unit of blood, elevated blood flow and oxygen delivery, and eventually a greater circulating hemoglobin mass. Acclimatization can be viewed as the end-stage process of how humans can best adjust to hypoxia. But optimal acclimatization takes from days to weeks, or perhaps even months.

The initial and immediate strategy to protect the body from hypoxia is to increase ventilation. This compensatory mechanism is triggered by stimulation of the carotid bodies, which sense hypoxemia (low arterial PO2), and increase central respiratory drive. This is a fast response, occurring within minutes of exposure to hypoxia persisting throughout high altitude exposure. This is why one cautions against the use of respiratory depressants such as alcohol and some sleeping medications, which can depress the hypoxic drive to breathe and may thus worsen hypoxemia. Pharmacological simulation of this natural process by acetazolamide, a respiratory stimulant and mild diuretic, largely protects from AMS and HACE by stimulating acclimatization. Circulatory responses are key to improving oxygen delivery, and are likely regulated by marked elevations in sympathetic activity. Field experience suggests that a marked elevation in early morning resting heart rate is a sign of challenges to acclimatization, perhaps secondary to increased hypoxemia, or dehydration. For the pathophysiology of AMS and HACE responses of the cerebral circulation are especially important. Maintenance of cerebral oxygen delivery is a critical factor for survival at high altitude. The balance between hypoxic vasodilation and hypocapnia-induced vasoconstriction determines overall cerebral blood flow (CBF). In a classic study, CBF increased 24% on abrupt ascent to 3810 m, and then returned to normal over 3 to 5 days. Recent studies, largely using regional transcranial Doppler measures of CBF velocity as a proxy for CBF, report discernible individual variation in the CBF response to hypoxia. All advanced brain imaging studies to date have shown both elevations in CBF in hypoxic humans and striking heterogeneity of CBF distribution in the hypoxic brain, with CBF rising up to 33% in the hypothalamus, and 20% in the thalamus with no other significant changes. Also, it is becoming clear that cerebral autoregulation, the process by which cerebral perfusion is maintained as blood pressure varies, is impaired in hypoxia. Thus, hypoxia modulates cerebral autoregulation and raises interesting questions about the importance of this process in AMS and acclimatization, since it appears to be a uniform response in all humans made hypoxemic. Further, hematocrit and hemoglobin concentration are elevated after 12 to 24 hours of hypoxic exposure due to a fall in plasma volume, but after several weeks,  plasma volume returns to near sea level values. Normalization of plasma volume coupled with an increase in red cell mass secondary to the hypoxia stimulated erythropoiesis leads to an increase in total blood volume after several weeks of acclimatization. Adequate iron stores are required for adequate hematologic acclimatization to high altitude. Acclimatization, then, is a series of physiological responses to hypoxia that serve to offset hypoxemia, improve systemic oxygen delivery, and avoid AMS and HACE. When acclimatization fails, or the challenge of hypoxia is too great, AMS and HACE can develop.

AMS occurs in susceptible individuals when ascent to high altitude outpaces the ability to acclimatize. For example, most people ascending very rapidly to high altitude will get AMS. The symptoms, although often initially incapacitating, usually resolve in 24 to 48 hrs. The incidence and severity of AMS depend on the rate of ascent and the altitude attained, the length of time at altitude, the degree of physical exertion, and the individual’s physiological susceptibility. The chief significance of AMS is that planned activities may be impossible to complete during the first few days at a new altitude due to symptoms. In addition, in a few individuals, AMS may progress to life-threatening HACE or HAPE. At 4000 m and above, the incidence of AMS ranges from 50% to 65% depending on the rate and mode of ascent, altitude reached, and sleeping altitude. A survey of 3158 travelers visiting resorts in the Rocky Mountains of Colorado revealed that 25% developed AMS, and most decreased their daily activity because of their symptoms.

Singh et al. proposed that the high-altitude syndromes are secondary to the body’s responses to hypobaric hypoxia, not due simply to hypoxemia. They based this conclusion on 2 observations:

  • there is a delay between the onset of hypoxia and the onset of symptoms after ascent (from hours to days), and
  • not all symptoms are immediately reversed with oxygen.

On the other hand, scientists have long assumed that AMS and HACE are due solely to hypoxia, based largely on 2 reports:

  • the pioneering experiments of Paul Bert and
  • the Glass House experiment of Barcroft.

When these assumptions were tested in a laboratory setting to study symptom responses to hypobaric hypoxia (simulated high altitude), hypoxia alone, and hypobaric normoxia, AMS occurred soonest and with greater severity with simulated altitude, compared with either normobaric hypoxia or normoxic hypobaria.  In 2 studies, one in normobaric hypoxia found no MRI signs of vasogenic edema but suggested that AMS was associated with “cytotoxic edema”, whereas a comparable study in hypobaric hypoxia found combined vasogenic and intracellular edema. The conclusions from the 2 studies have very different implications for refining a theory of the pathophysiology of AMS. Although the studies were not designed for a direct comparison between hypobaria and hypoxia, the discrepancy points out an assumption about normobaric hypoxia and the pathophysiology of AMS that may warrant further investigation.

Our central hypothesis regarding the pathophysiology of AMS, and by extension of HACE, is that it is centered on dysfunction within the brain. This is not a new idea, but it is one of current intense interest thanks to advances in brain imaging and neuroscience techniques. Barcroft, writing in 1924, argued that the brain’s response to hypoxia was central to understanding the pathophysiology of mountain sickness.

A low ventilatory response to hypoxia coupled with increased symptoms of AMS led to intensive investigation of a link between the chemical control of ventilation and the pathogenesis of AMS. The results of these investigations suggest that for most people, the ventilatory response to hypoxia has little predictive value for AMS risk. Only if the extremes of ventilator responsiveness are contrasted can accurate predictions be made, where those with extremely low ventilatory drives being more likely to suffer AMS. At the extreme end of the distribution (i.e., for very high responses), the protective role of a brisk hypoxic ventilatory response may be due to increased arterial oxygen content and cerebral oxygen delivery despite mild hypocapnic cerebral vasoconstriction.

Hansen and Evans were the first to publish a comprehensive hypothesis of the pathophysiology of AMS centered on the brain. Their theory posited that compression of the brain, either by increased cerebral venous volume, reduced absorption of cerebral spinal fluid, or increased brain-tissue hydration (edema), initiates the development of the symptoms and signs of AMS and HACE. Ross built on these ideas with his “tight fit hypothesis,” published in 1985, and others have developed these ideas into a series of testable hypotheses congruent with today’s knowledge of AMS and HACE. The tight fit hypothesis states that expanded intracranial volume (due to the reasons put forth by Hansen and Evans, or other causes) plus the volume available for intracranial buffering of that expanded volume would predict who would get AMS. Greater buffering capacity leads to AMS resistance, lower buffering capacity, or a ‘tight fit’ of the brain in the cranial vault, would lead to greater AMS susceptibility. Overall, it is clear that brain volume increases in humans on exposure to hypoxia. It is less certain whether this elevation in brain volume plays a role in AMS.

Hackett’s pioneering MRI study in HACE, with marked white matter edema suggestive of a vasogenic origin, has led to a decade of studies looking for a similar finding in AMS. In moderate to severe AMS, all imaging studies have shown some degree of cerebral edema. But in mild to moderate AMS, admittedly an arbitrary and subjective distinction, brain edema is present in some MRI studies of AMS subjects, but not in all. It seems reasonable to conclude from the available data that the increase in brain volume observed is at least partially due to brain edema, and that earlier studies missed the edema more for technical than physiological reasons. It is less clear whether the brain edema is largely of intracellular or vasogenic origin, and what role if any it plays in the pathophysiology of AMS.

Although we support transcranial doppler for many investigations in integrative physiology, the complex interplay of hypoxia and hypocapnia that is present in acutely hypoxic humans may present a situation where whole brain imaging is a more reliable and accurate tool to discern the role of CBF in the onset of AMS. To date, no brain imaging studies have addressed global cerebral perfusion in AMS.

The management of AMS and HACE is based on our current understanding of the physiological and pathophysiological responses to hypoxia. Hypoxia itself, however, does not immediately lead to AMS as there is a delay of several hours after arrival at high altitude before symptoms develop. Increased knowledge of hypoxic inducible factor and cytokines that alter capillary permeability may lead to the discovery of new drugs for the prevention and alleviation of AMS and HACE.

Much work has focused on the role of vascular endothelial growth factor (VEGF), a potent permeability factor up-regulated by hypoxia. Some studies have found no evidence of an association of changes in plasma concentrations of VEGF and AMS, whereas others support the hypothesis that VEGF contributes to the pathogensis of AMS. Clearly a better understanding of the mechanisms of increased capillary permeability of cerebral capillaries will greatly enhance the management of AMS and HACE.

Flying high: A theoretical analysis of the factors limiting exercise performance in birds at altitude

Graham R. Scott, William K. Milsom
Respiratory Physiology & Neurobiology 154 (2006) 284–301
http://dx.doi.org:/10.1016/j.resp.2006.02.012

The ability of some bird species to fly at extreme altitude has fascinated comparative respiratory physiologists for decades, yet there is still no consensus about what adaptations enable high altitude flight. Using a theoretical model of O2 transport, we performed a sensitivity analysis of the factors that might limit exercise performance in birds. We found that the influence of individual physiological traits on oxygen consumption (˙VO2 ) during exercise differed between sea level, moderate altitude, and extreme altitude. At extreme altitude, hemoglobin (Hb) O2 affinity, total ventilation, and tissue diffusion capacity for O2 (DTO2) had the greatest influences on VO2; increasing these variables should therefore have the greatest adaptive benefit for high altitude flight. There was a beneficial interaction between DTO2 and the P50 of Hb, such that increasing DTO2 had a greater influence on VO2 when P50 was low. Increases in the temperature effect on P50 could also be  beneficial for high flying birds, provided that cold inspired air at extreme altitude causes a substantial difference in temperature between blood in the lungs and in the tissues. Changes in lung diffusion capacity for O2, cardiac output, blood Hb concentration, the Bohr coefficient, or the Hill coefficient likely have less adaptive significance at high altitude. Our sensitivity analysis provides theoretical suggestions of the adaptations most likely to promote high altitude flight in birds and provides direction for future in vivo studies.

The bird lung is unique among the lungs of air-breathing vertebrates, with a blood flow that is crosscurrent to gas flow, and a gas flow that occurs unidirectionally through rigid parabronchioles. As such, bird lungs are inherently more efficient than the lungs of other air-breathing vertebrates (Piiper and Scheid, 1972, 1975). While this may partially account for the greater hypoxia tolerance of birds in general when compared to mammals (cf. Scheid, 1990), its presence in all birds excludes the crosscurrent lung as a possible adaptation specific to high altitude fliers. Similarly, an extremely small diffusion distance across the blood–gas interface compared to other air breathers seems to be a characteristic of all bird lungs, and not just those of high fliers (Maina and King, 1982; Powell and Mazzone, 1983; Shams and Scheid, 1989). Partly because of this small diffusion distance, the inherent O2 diffusion capacity across the gas–blood interface (DLO2) is generally high in birds. Interestingly, pulmonary vasoconstriction does not appear to increase during hypoxia in bar-headed geese (Faraci et al., 1984a). This may be a significant advantage during combined exercise and severe hypoxia, and suggests that regulation of lung blood flow could be important in high altitude birds. In addition, the CO2/pH sensitivity of ventilation is commonly assessed by comparing the isocapnic and poikilocapnic hypoxic ventilatory responses; however, the isocapnic ventilatory responses to hypoxia of both low and high altitude birds have not been compared. In this regard, the ventilator response in high altitude birds may also depend on their capacity to maintain intracellular pH during alkalosis, or to buffer changes in extracellular pH due to hyperventilation. It therefore remains to be conclusively determined whether high altitude fliers have a greater capacity to increase ventilation during severe hypoxia.

After diffusing into the blood in the lungs, oxygen is primarily circulated throughout the body bound to hemoglobin. A high cardiac output is therefore important for exercise at high altitude to supply the working muscle with adequate amounts of O2. Indeed, animals selectively bred for exercise performance have higher maximum cardiac outputs, as do species that have evolved for exercise performance. Whether cardiac output limits exercise performance per se, however, is less clear; other factors may limit intense exercise, and in more athletic species (or individuals) cardiac output may be higher simply out of necessity. Excessive cardiac output may even be detrimental if blood transit times in the lungs or tissues are substantially reduced. Unfortunately, very little is known about cardiac performance in high flying birds. Both the high altitude bar-headed goose and the low altitude pekin duck can increase cardiac output at least five-fold during hypoxia at rest (Black and Tenney, 1980), but no comparison of maximum cardiac performance has been made between high and low altitude birds.

Once oxygenated blood is circulated to the tissues, O2 moves to the tissue mitochondria, the site of oxidative phosphorylation and oxygen consumption. Transport of oxygen from the blood to the mitochondria involves several steps. Oxygen must first dissociate from Hb and diffuse through the various compartments of the blood, but in both birds and mammals the conductances of these steps are high, and are unlikely to impose much of a limitation to O2 transport. In contrast, diffusion across the vascular wall and through the extracellular spaces is thought to provide the most sizeable limitation to O2 transport. Consequently, the size of the capillary–muscle fiber interface is an extremely important determinant of a muscle’s aerobic capacity. Finally, oxygen diffuses across the muscle fiber membrane and moves through the cytoplasm until it associates with cytochrome c oxidase, the O2 acceptor in the mitochondrial electron transport chain. Myoglobin probably assists intracellular O2 transport, so diffusion through the muscle likely provides very little resistance to O2 flux.

It is obvious that the ability of some bird species to fly at extreme altitudes is poorly understood. The adaptive benefit of high hemoglobin oxygen affinity is well established, but its relative importance is unknown. Some evidence suggests that traits increasing oxygen diffusion capacity in flight muscle are adaptive in high fliers as well, but the adaptive significance of differences in the respiratory and cardiovascular systems of high altitude fliers is not clear. The remainder of this study assesses these possibilities using theoretical sensitivity analysis, and explores potential adaptations for high altitude flight in birds.

Oxygen transport in birds

Oxygen transport in birds

Oxygen transport in birds. The crosscurrent parabronchial lung is unidirectionally ventilated by air sacs, and oxygen diffuses into blood capillaries from air capillaries (not shown) all along the length of the parabronchi. Oxygen is then circulated in the blood, and diffuses to mitochondria in the tissues. The rate of oxygen transport at both the lungs and tissues can be calculated using the Fick equation, and the amount of O2 transferred from the lungs into the blood can be calculated using an oxygen conservation equation.

Oxygen tensions in the lung

Oxygen tensions in the lung

Oxygen tensions in the lung (A) and tissue (B) capillaries during normoxia. In the crosscurrent avian lung, PO2 varies in two dimensions: PO2 increases along the path of blood flow through the lungs, but does not increase by as much at the end of the parabronchi as at the start (gas PO2 decreases along the length of the parabronchi). In the tissues, blood PO2 decreases continuously along the capillary length as O2 diffuses to tissue mitochondria. To reach a solution, our model iterates between gas transport calculations in the lungs (A) and tissues (B) until a stable result is reached.

varying different biochemical features of hemoglobin (Hb) on oxygen consumption

varying different biochemical features of hemoglobin (Hb) on oxygen consumption

The effects of varying different biochemical features of hemoglobin (Hb) on oxygen consumption during exercise in normoxia (PIO2 of 150 Torr; red), moderate hypoxia (84 Torr; green dashed), and severe hypoxia (30 Torr; dark blue). (A) P50, the PO2 at 50% Hb saturation; (B and C) Bohr coefficient (φ); and (D and E) Hill coefficient (n) (see Section 2 for a mathematical description of each). In (B)–(E), the effects of each variable were assessed at the P50 of pekin ducks (40 Torr; B and D) as well as the P50 of bar-headed geese (25 Torr; C and E).

Unlike in vivo studies, theoretical sensitivity analyses allow individual physiological variables to be altered independently so their individual effects on oxygen consumption can be assessed. By applying this analysis to hypoxia in birds, we feel we can predict which factors most likely limit oxygen consumption and exercise performance. As a consequence, our analysis identifies which steps in the oxygen cascade can provide the basis for adaptive change in birds that evolved for high altitude flight, namely ventilation and tissue diffusion capacity.

Since our interest was in the factors limiting exercise performance at altitude, the starting data for our model were obtained from previous studies on pekin ducks near maximal oxygen consumption. These ducks were exercising on a treadmill, however, and were not flying. Unfortunately, to the best of our knowledge only one previous study has made all the required measurements for this analysis during flight, and this was only done in normoxia (in pigeons, Butler et al., 1977). Pekin ducks are the only species for which we could find all the required measurements for our analysis during exercise in both normoxia and hypoxia. Only the lung and tissue diffusion capacities remained to be calculated in our analysis, but previous experimental determinations of DLO2 in pekin ducks were similar to the values calculated in this study (Scheid et al., 1977). Similar values for DTO2 are not available.

The physiological variables limiting exercise performance in birds during moderate hypoxia are similar to those limiting performance in normoxia. DTO2 continues to pose the greatest limitation, and limitations imposed by the circulation (˙Q and CHb) are still greater at a lower P50. Unlike normoxia, however, ˙VO2 in moderate hypoxia appears to be limited less by the circulation and more by respiratory variables, as is also the case in humans (Wagner, 1996). The most substantial difference between severe hypoxia and normoxia/moderate hypoxia is in the effects of altering ventilation. Ventilation appears to become a major limitation to exercise performance at extreme altitude. DTO2 also appears to limit ˙VO2 in severe hypoxia, but only at lower P50 values. This is not entirely unsurprising: in severe hypoxia the venous blood of pekin ducks (a species which has a higher P50) is almost completely deoxygenated in vivo, so there are no possible benefits of increasing DTO2 . At the lower P50, there is a substantially higher arterial oxygen content, so more oxygen can be removed, and increasing DTO2 can have a greater influence. In humans during severe hypoxia, DTO2, DLO2, and ˙V have the greatest influence on exercise performance.

Tissue diffusion capacity should also be adaptive in high altitude birds with a high hemoglobin O2 affinity. In the present study, a simultaneous decrease in P50 (from 40 to 25 Torr) and increase in DTO2 (twofold) increased ˙VO2 by 51%. Thus, in high flying birds that are known to have a low P50, such as the barheaded goose and Ruppell’s griffon (Gyps rueppellii), increases in flight muscle diffusion capacity should be of extreme importance. This suggestion is supported by research demonstrating greater muscle capillarization in bar-headed geese than in low altitude fliers, as the size of the capillary–muscle fiber interface is known to be the primary structural determinant of O2 flux into the muscle.

Our analysis suggests that an enhanced capacity to increase ventilation should also benefit birds significantly in severe hypoxia, and could therefore be an important source of adaptation for high altitude flight. This is likely true regardless of P50; although there is a small amount of interaction between P50 and ventilation, increasing ˙V always had a substantial effect on oxygen consumption. Data from the literature addressing this possibility have unfortunately been inconclusive. Both bar-headed geese and pekin ducks can effectively increase ventilation, thus reducing the inspired-arterial O2 difference, during severe poikilocapnic hypoxia at rest, as well as during moderate poikilocapnic hypoxia and running exercise.

oxyhemoglobin dissociation curve

oxyhemoglobin dissociation curve

In contrast to the Bohr effect and Hill coefficient, the temperature effect on Hb-O2 binding affinity may have a substantial effect on oxygen consumption, and may therefore be a source of adaptive change for high altitude flight. An effect of temperature on ˙VO2 may arise if hyperventilation during flight at extreme altitude cools the pulmonary blood. This would reduce the P50 of Hb in the lungs, and thus facilitate oxygen uptake. When this blood enters the exercising muscles it would then be rewarmed to body temperature, and oxygen would be released from Hb. Our modelling suggests that a temperature effect on Hb could significantly enhance ˙VO2 . The greater the difference in temperature between blood in the lungs and in the muscles, and the greater the temperature effect on Hb-O2 binding, the greater the increase in ˙VO2 . At normal levels of temperature sensitivity, the increase in ˙VO2 was approximately 5% for every 1 ◦C difference. It could be adaptive at high altitude to alter the magnitude of the temperature effect on Hb while allowing lung temperature to fall. At present, however, it is unknown whether the Hb of high altitude birds has a heightened sensitivity to temperature, or whether pulmonary blood is actually cooled during high altitude flight.

Using a theoretical sensitivity analysis that allows individual physiological variables to be altered independently, we have identified the factors most likely to limit oxygen consumption and exercise performance in birds, and by extension, the physiological changes that are likely adaptive for high altitude flight. The adaptive benefits of some of these changes, in particular hemoglobin oxygen affinity, are already well established for high flying birds. For other traits, such as an enhanced hypoxic ventilatory response or O2 diffusion capacity of flight muscle, adaptive differences have not been conclusively recognized in studies in vivo. Furthermore, the beneficial interaction between increasing DTO2 and decreasing hemoglobin P50 has not yet been demonstrated in vivo. Our theoretical analysis suggests that changes in these respiratory processes could also adapt birds to environmental extremes, and future studies should explore these findings.

Adaptation and Convergent Evolution within the Jamesonia-Eriosorus Complex in High-Elevation Biodiverse Andean Hotspots

Patricia Sanchez-Baracaldo, Gavin H. Thomas
PLoS ONE 9(10): e110618. http://dx.doi.org:/10.1371/journal.pone.0110618

The recent uplift of the tropical Andes (since the late Pliocene or early Pleistocene) provided extensive ecological opportunity for evolutionary radiations. We test for phylogenetic and morphological evidence of adaptive radiation and convergent evolution to novel habitats (exposed, high-altitude paramo habitats) in the Andean fern genera Jamesonia and Eriosorus. We construct time-calibrated phylogenies for the Jamesonia-Eriosorus clade. We then use recent phylogenetic comparative methods to test for evolutionary transitions among habitats, associations between habitat and leaf morphology, and ecologically driven variation in the rate of morphological evolution. Paramo species (Jamesonia) display morphological adaptations consistent with convergent evolution in response to the demands of a highly exposed environment but these adaptations are associated with microhabitat use rather than the paramo per se. Species that are associated with exposed microhabitats (including Jamesonia and Eriorsorus) are characterized by many but short pinnae per frond whereas species occupying sheltered microhabitats (primarily Eriosorus) have few but long pinnae per frond. Pinnae length declines more rapidly with altitude in sheltered species. Rates of speciation are significantly higher among paramo than non-paramo lineages supporting the hypothesis of adaptation and divergence in the unique Pa´ramo biodiversity hotspot.

AltitudeOmics: Rapid Hemoglobin Mass Alterations with Early Acclimatization to and De-Acclimatization from 5,260 m in Healthy Humans

Benjamin J. Ryan, NB Wachsmuth, WF Schmidt, WC Byrnes, et al.
PLoS ONE 9(10): e108788. http://dx.doi.org:/10.1371/journal.pone.0108788

It is classically thought that increases in hemoglobin mass (Hb mass) take several weeks to develop upon ascent to high altitude and are lost gradually following descent. However, the early time course of these erythropoietic adaptations has not been thoroughly investigated and data are lacking at elevations greater than 5,000 m, where the hypoxic stimulus is dramatically increased. As part of the AltitudeOmics project, we examined Hb mass in healthy men and women at sea level (SL) and 5,260 m following 1, 7, and 16 days of high altitude exposure (ALT1/ALT7/ALT16). Subjects were also studied upon return to 5,260 m following descent to 1,525 m for either 7 or 21 days. Compared to SL, absolute Hb mass was not different at ALT1 but increased by 3.7-5.8% (mean 6 SD; n = 20; p<0.01) at ALT7 and 7.6-6.6% (n = 21; p=0.001) at ALT16. Following descent to 1,525 m, Hb mass was reduced compared to ALT16 (-6.0+3.7%; n = 20; p = 0.001) and not different compared to SL, with no difference in the loss in Hb mass between groups that descended for 7 (-6.3+3.0%; n = 13) versus 21 days (-5.7+5.0; n = 7). The loss in Hb mass following 7 days at 1,525 m was correlated with an increase in serum ferritin
(r =20.64; n = 13; p,0.05), suggesting increased red blood cell destruction. Our novel findings demonstrate that Hb mass increases within 7 days of ascent to 5,260 m but that the altitude-induced Hb mass adaptation is lost within 7 days of descent to 1,525 m. The rapid time course of these adaptations contrasts with the classical dogma, suggesting the need to further examine mechanisms responsible for Hb mass adaptations in response to severe hypoxia.

Cardiovascular adjustments for life at high altitude

Roger Hainsworth, Mark J. Drinkhill
Respiratory Physiology & Neurobiology 158 (2007) 204–211
http://dx.doi.org:/10.1016/j.resp.2007.05.006

The effects of hypobaric hypoxia in visitors depend not only on the actual elevation but also on the rate of ascent. There are increases in sympathetic activity resulting in increases in systemic vascular resistance, blood pressure and heart rate. Pulmonary vasoconstriction leads to pulmonary hypertension, particularly during exercise. The sympathetic excitation results from hypoxia, partly through chemoreceptor reflexes and partly through altered baroreceptor function. Systemic vasoconstriction may also occur as a reflex response to the high pulmonary arterial pressures. Many communities live permanently at high altitude and most dwellers show excellent adaptation although there are differences between populations in the extent of the ventilatory drive and the erythropoiesis. Despite living all their lives at altitude, some dwellers, particularly Andeans, may develop a maladaptation syndrome known as chronic mountain sickness. The most prominent characteristic of this is excessive polycythemia, the cause of which has been attributed to peripheral chemoreceptor dysfunction. The hyperviscous blood leads to pulmonary hypertension, symptoms of cerebral hypoperfusion, and eventually right heart failure and death.

High altitude places are not only destinations of adventurous travelers, many people are born, live their lives and die in these cold and hypoxic regions. According to WHO, in 1996 there were approximately 140 million people living at altitudes over 2,500m and there are several areas of permanent habitation at over 4,000 m. These are in three main regions of the world: the Andes of South America, the highlands of Eastern Africa, and the Himalayas of South-Central Asia. This review is concerned with the effects of exposure to high altitude on the cardiovascular system and its autonomic control, in visitors, and the means by which the permanent high altitude dwellers have adapted to their environment.

For visitors the period of initial adaptation, i.e. the first days and weeks following arrival at attitude, is a critical time since it is during this period that acute mountain sickness and/or pulmonary edema may occur. The processes of adaptation occurring during this initial period may well determine the individual’s ability to continue to function normally. Recent studies in animals and man have highlighted the role of the autonomic nervous system in adaptation and in particular the importance of sympathetic activation of the cardiovascular system following high altitude exposure.

An increase in resting heart rate in response to acute hypoxia has been
described in several species including man. Vogel and Harris (1967)
investigated the effects of simulated exposure to high altitude in man
at pressures equivalent to 600, 3,400 and 4,600m using a hypobaric
chamber. Each level of chamber pressure was developed over a 30 min
period andwas maintained for 48 h in an attempt to simulate expedition
conditions. After 10 h at the equivalent of 3,400 m resting
heart rate was significantly increased and by 40 h it had increased by
16% from the resting value at 600 m. At 4,600 m it increased by 34%.
Similar findings, an increase in heart rate of 18%, were shown following
ascent to 4,300 m for periods up to 5 weeks. However, this study also
demonstrated that the rate of ascent also influenced the magnitude of
the heart rate increase. A gradual increase in altitude over a period
of 2 weeks resulted in the resting heart rate increasing by 25%
compared with an abrupt ascent which resulted in an increase of
only 9%. As subjects acclimatize at altitudes up to about 4,500 m
much of the increase in heart rate is lost and resting heart rates
return towards their sea level values. Acute hypoxia also causes
increases in cardiac output both at rest and for given levels of
exercise compared with values during normoxia.

The effect of hypoxia on the pulmonary circulation is dramatic
resulting in pulmonary hypertension caused by an increase in
pulmonary vascular resistance. The onset has been shown in man
to be very rapid, reaching a maximum within 5 min. Zhao et al.
(2001) demonstrated that breathing 11% oxygen for 30 min
increased mean pulmonary artery pressure by 56%, from 16 to
25 mmHg. The effect of hypoxia on the pulmonary circulation is
even more pronounced during exercise, as demonstrated in studies
carried out on subjects of Operation Everest II. Resting pulmonary
artery pressure increased from 15 mmHg at sea level to 34 mmHg
at the equivalent of 8,840 m. During near maximal exercise at
8,840 m it increased from the sea level value of 33–54 mm Hg.
In the short term the mechanism of this pulmonary artery vaso-
constriction has been shown to involve inhibition of O2 sensitive
K+ channels leading to depolarization of pulmonary artery smooth
muscle cells and activation of voltage gated Ca2+ channels. This
causes Ca2+ influx and vasocon-striction. This process is
immediately reversed by breathing oxygen.

Healthy high altitude residents show excellent adaptation to their
environment. These adaptations are likely to be associated with
altered gene expression as the expression of genes associated with
vascular control and reactions to hypoxia have been found to be high
in altitude dwellers. Different communities, however, seem to adopt
different adaptation strategies. For example Andeans hyperventilate
to decrease end-tidal and arterial CO2 levels to as low as 25 mmHg
and have hemoglobin levels well above those in sea-level people.
Tibetans Hyperventilate but have normal hemoglobin levels below
4,000 m. Ethiopian highlanders, on the other hand, have CO2 and
hemoglobin levels similar to those of sea-level dwellers.

Blood volumes are larger in high altitude dwellers. In Andeans this
is due to large packed cell volumes whereas in Ethiopians it was the
plasma volumes that were large. Probably as the result of the large
blood volumes, tolerance to orthostatic stress was greater than that
in sea-level residents.

CMS is a condition frequently found in long term residents of high
altitudes, particularly in the Andes where it is a major public health
problem. It also occurs in residents on the Tibetan plateau, although
not in Ethiopians. Patients with CMS develop excessive polycythemia
and various clinical features including dyspnea, palpitations, insomnia,
dizziness, headaches, confusion, loss of appetite, lack of mental
concentration and memory alterations. Patients may also complain
of decreased exercise tolerance, bone pains, acral paresthesia and
occasionally hemoptysis. The impairment of mental function may
be reversed by phlebotomy. Physical examination reveals cyanosis,
due to the combination of polycythemia and low oxygen saturation,
and a marked pigmentation of the skin exposed to the sun.
Hyperemia of conjunctivae is characteristic and the retinal vessels
are also dilated and engorged. The second heart sound is frequently
accentuated and there is an increased cardiac size, mainly due to
right ventricular hypertrophy. As the condition progresses, overt
congestive heart failure becomes evident, characterized by dyspnea
at rest and during mild effort, peripheral edema, distension of
superficial veins, and progressive cardiac dilation.

The major mechanism for the control of blood pressure is through
regulation of peripheral vascular resistance, but most studies have
examined only the control of heart rate. We have recently studied
the responses of forearm vascular resistance to carotid baroreceptor
stimulation in high altitude residents with and without CMS, both at
their resident altitude and shortly after descent to sea level. Results
showed that baroreflex “set point” was higher in CMS, but only at
altitude. At sea level, values were similar.

The chronic hypoxia at high altitude stresses many of the body’s
homeostatic mechanisms. There have been many investigations
which have examined the effects on respiration. However, cardio-
vascular effects are no less important and it is largely through effects
on the cardiovascular system that both acute and chronic mountain
sickness are caused. The hypoxia exerts both direct and reflex effects.
In the lung it causes vasoconstriction and pulmonary hypertension.
The sympathetic nervous system is excited partly through a central
effect of the hypoxia, through stimulation of chemoreceptors and
possibly pulmonary arterial baroreceptors and altered systemic
baroreceptor function. In some individuals the excessive hemopoiesis
causes increased blood viscosity and tissue hypoperfusion leading
to the syndrome of chronic mountain sickness.

New Insights in the Pathogenesis of High-Altitude Pulmonary Edema

Urs Scherrer, Emrush Rexhaj, Pierre-Yves Jayet, et al.
Progress in Cardiovascular Diseases 52 (2010) 485–492
http://dx.doi.org:/10.1016/j.pcad.2010.02.004

High-altitude pulmonary edema is a life-threatening condition occurring in predisposed but otherwise healthy individuals. It therefore permits the study of underlying mechanisms of pulmonary edema in the absence of confounding factors such as coexisting cardiovascular or pulmonary disease, and/or drug therapy. There is evidence that some degree of asymptomatic alveolar fluid accumulation may represent a normal phenomenon in healthy humans shortly after arrival at high altitude. Two fundamental mechanisms then determine whether this fluid accumulation is cleared or whether it progresses to HAPE: the quantity of liquid escaping from the pulmonary vasculature and the rate of its clearance by the alveolar respiratory epithelium. The former is directly related to the degree of hypoxia induced pulmonary hypertension, whereas the latter is determined by the alveolar epithelial sodium transport. Here, we will review evidence that, in HAPE-prone subjects, impaired pulmonary endothelial and epithelial NO synthesis and/or bioavailability may represent a central underlying defect predisposing to exaggerated hypoxic pulmonary vasoconstriction and, in turn, capillary stress failure and alveolar fluid flooding. We will then demonstrate that exaggerated pulmonary hypertension, although possibly a condition sine qua non, may not always be sufficient to induce HAPE and how defective alveolar fluid clearance may represent a second important pathogenic mechanism.

Cerebral Blood Flow at High Altitude

Philip N. Ainslie and Andrew W. Subudhi
High Altitude Medicine & Biology 2014; 15(2): 133–140
http://dx.doi.org:/10.1089/ham.2013.1138

This brief review traces the last 50 years of research related to cerebral blood flow (CBF) in humans exposed to high altitude. The increase in CBF within the first 12 hours at high altitude and its return to near sea level values after 3–5 days of acclimatization was first documented with use of the Kety-Schmidt technique in 1964. The degree of change in CBF at high altitude is influenced by many variables, including arterial oxygen and carbon dioxide tensions, oxygen content, cerebral spinal fluid pH, and hematocrit, but can be collectively summarized in terms of the relative strengths of four key integrated reflexes:

  • hypoxic cerebral vasodilatation;
  • 2) hypocapnic cerebral vasoconstriction;
  • 3) hypoxic ventilatory response; and
  • 4) hypercapnic ventilatory response.

Understanding the mechanisms underlying these reflexes and their interactions with one another is critical to advance our understanding of global and regional CBF regulation. Whether high altitude populations exhibit cerebrovascular adaptations to chronic levels of hypoxia or if changes in CBF are related to the development of acute mountain sickness are currently unknown; yet overall, the integrated CBF response to high altitude appears to be sufficient to meet the brain’s large and consistent demand for oxygen.

Relative to its size, the brain is the most oxygen dependent organ in the body, but many pathophysiological and environmental processes may either cause or result in an interruption to its oxygen supply. As such, studying the brain at high altitude is an appropriate model to investigate both acute and chronic effects of hypoxemia on cerebrovascular function. The cerebrovascular responses to high altitude are complex, involving mechanistic interactions of physiological, metabolic, and biochemical processes.

This short review is organized as follows: An historical overview of the earliest CBF measurements collected at high altitude introduces a summary of reported CBF changes at altitude over the last 50 years in both lowlanders and high-altitude natives. The most tenable candidate mechanism(s) regulating CBF at altitude are summarized with a focus on available data in humans, and a role for these mechanisms in the pathophysiology of AMS is considered. Finally, suggestions for future directions are provided.

Angelo Mosso (1846–1910) is undoubtedly the forefather of high altitude cerebrovascular physiology. In order to pursue his principal curiosity of the physiological effects of hypobaria, Mosso built barometric chambers and was reported to expose himself pressures as low as 192 mmHg (equivalent to > 10,000 m). He was also responsible for the building of the Capanna Margherita laboratory on Monta Rosa at 4,559 m. In both settings, Mosso utilized his hydrosphygmomanometer to measure changes in ‘‘brain pulsations’’ in patients that had suffered removal of skull sections, due to illness or trauma. Indicative of changes in CBF, these recordings preceded the next estimates of CBF in humans by some 50 years.

At sea level, Kety and Schmidt (1945) were the first to quantify human CBF using an inert tracer (nitrous oxide, N2O) combined with arterial and jugular venous sampling. This method for the measurement of global CBF is based on the Fick principle, whereby the integrated difference of multiple arterial and venous blood samples during the first 10 or more minutes after the sudden introduction into the lung of a soluble gas tracer is inversely proportional to cerebral blood flow.  In 1948, they showed that breathing 10% oxygen increased CBF by 35%; however, it was not until 1964 that the first measurements of CBF were made in humans at high altitude. The motivation for these high altitude experiments was stimulated, in part, from the earlier discovery of the brain’s ventral medullary cerebrospinal fluid (CSF) pH sensors in animals. Following the location of these central chemoreceptors, Severinghaus and colleagues examined in humans the role of CSF pH and bicarbonate in acclimatization to high altitude (3,810 m) at the White Mountain (California, USA) laboratories (Severinghaus et al., 1963). A year later, at the same location, John Severinghaus performed his seminal study of CBF at high altitude. He was joined by Tom Hornbein—shortly after his first ascent of Everest by the West Ridge—who was part of the research team and also volunteered for the study (Fig.). The results showed clear time dependent changes in CBF during acclimatization to high altitude (HA).

the Kety-Schmidt nitrous oxide method of measuring CBF

the Kety-Schmidt nitrous oxide method of measuring CBF

  • From left to right, Larry Saidman (administering the gas), Tom Hornbien (volunteer), Ed Munson (drawing jugular venous blood samples), and John Severinghaus. Here (1964) the Kety-Schmidt nitrous oxide method of measuring CBF is used. The subject breathed about 15% N2O for 15 min while arterial and jugular venous blood was frequently sampled. (B) Results from Severinghaus et al. (1966). Graphs shows that CBF as estimated by cerebral A-VO2 differences from sea level controls increased about 24% within hours of arrival at 3810 m, and fell over 4 days to about 13% above control. CBF by the N2O method was increased by 40% on day 1, and returned to 6% above control on day 4. However, the N2O method data had greater variance. Acute normoxia on day 1 and day 4 returned CBF to sea level values within 15 min. Photograph courtesy of Dr. John W Severinghaus.

Native Tibetan (or Himalayan) and Andean populations arrived approximately 25,000 and 11,000 years ago, suggesting that these populations either carried traits that allowed them to thrive at high altitude or were able to adapt to the environment. The physiological and genetic traits associated with native high-altitude populations have been elegantly reviewed (Beall, 2007; Erzurum et al., 2007; Frisancho, 2013). As such, this topic is briefly summarized here with the focus on CBF at altitude in context of Andean and Tibetan high-altitude residents.

In general, native Andeans have lower CBF values compared to sea level natives. The first evidence suggesting lower flow was reported in 8 Peruvian natives living at 4300m altitude in Cerro de Pasco (Milledge and Sørensen, 1972). The authors found the mean arterial–venous oxygen content difference across the brain was 7.9 – 1 vol%, about 20% higher than the published sea level mean of 6.5 vol%. They suggested that CBF probably was proportionately about 20% below sea level normal values, assuming that brain metabolic rate was normal, and postulated that the mechanism might be high blood viscosity given the high hematocrit (58 – 6%) in these subjects. However, since the cerebral metabolic rate for oxygen (CMRO2) is constant even in severe hypoxia (Kety and Schmidt 1948b; Ainslie et al. 2013), the inverse linear relationship between CBF and arterial–venous oxygen content differences could also explain the reduction in CBF, as less flow would be needed to match the oxygen demand of the brain when arterial content is elevated. A similar study (Sørensen et al., 1974), using arterio-venous differences combined (in a subgroup) with a modified version of Kety–Schmidt method (krypton instead of N2O,) conducted in high-altitude residents in La Paz in Bolivia at 3800 m, also reported a 15%–20% reduction in CBF (with a reported average hematocrit of 50%) compared to a sea level control group.

Percent changes in cerebral blood flow

Percent changes in cerebral blood flow

Percent changes in cerebral blood flow (D%CBF, graph A), arterial oxygen content (Cao2, graph B), and cerebral oxygen delivery (CDO2, graph C) with time at high-altitude from seven studies at various altitudes and durations. Severinghaus et al. (1966) studied CBF using the Kety-Schmidt technique in five subjects brought rapidly by car to 3810 m. Using the Xe133 method, Jensen et al. (1990) measured CBF in 12 subjects at 3475 m. Huang et al. (1987) measured ICA and VA blood velocities as a metric of CBF on Pikes Peak (4300 m). Baumgartner et al. (1994) studied 24 subjects who rapidly ascended to 3200m by cable car, slept one night at 3600 m, and ascended by foot to 4559m the next day. Cerebral blood flow was estimated by transcranial Doppler ultrasound. About two-thirds of the subjects developed symptoms of AMS, data included are the mean of all subjects. Lucas et al. (2011) employed an 8–9 day ascent to 5050m and estimated changes in CBF by transcranial Doppler ultrasound of the middle cerebral artery. Willie et al. (2013) following the same ascent measured flow (Duplex ultrasound; and TCCD) in the ICA and VA and estimated global flow from: 2*ICA + 2* VA. The same methodological approach was used time Subudhi upon rapid ascent via car and oxygen breathing to 5240 m (Subudhi et al. 2013). Cao2 was calculated from: (1.39 · [Hb] · SaO2) + Pao2 *0.003. In some studies [Hb] data were not available, and typical data from previous studies over comparable time at related elevation were used. In other studies, Pao2 was not always reported; therefore, Sao2 was used to estimate Pao2 via (Severinghaus, 1979).

Only two studies have measured serial changes in CBF during progressive ascent to high altitude, but the findings may help explain small discrepancies between studies. In 2011, Wilson et al. (2011) measured diameter and velocity in the MCA (using transcranial color-coded Duplex-ultrasound, TCCD) following partial acclimation to 5300m (n = 24), 6400 m (n = 14), and 7950m (n = 5). Remarkable elevations (200%) in flow in the MCA occurred at 7950 m. Notably, the authors estimated *24% dilation of the MCA occurred at 6400 m. Dilation of the MCA further increased to *90% at 7950m (Fig.) and was rapidly reversed with oxygen supplementation (Fig.). Cerebral oxygen delivery and oxygenation were maintained by commensurate elevations of CBF even at these extreme altitudes. In another recent study, CBF and MCA diameter were measured at 1338 m, 3440 m, 4371 m, and over time at 5050 m (Willie et al., 2013). Dilation of the MCA was observed upon arrival at 5050 m with subsequent normalization of CBF and MCA diameter by days 10–12. Such findings are consistent with unchanged diameter following 17 days at 5400m (Wilson et al., 2011). It is important to note that according to Poiseuille’s Law, flow is proportional to radius raised to the fourth power. Therefore, consistent with previous concerns about TCD (Giller, 2003), that the MCA dilates at such levels of hypoxemia indicates that previous studies using TCD at altitude may have underestimated flow (see previous Fig.) and thus may explain differences between studies. These findings are particularly important because they suggest regional regulation of CBF occurs in both large and small cerebral arteries.

Changes in blood flow in the middle cerebral artery (MCA) upon progressive ascent to 7950 m

Changes in blood flow in the middle cerebral artery (MCA) upon progressive ascent to 7950 m

Changes in blood flow in the middle cerebral artery (MCA) upon progressive ascent to 7950 m. Data were collected following partial acclimation to 5300 m (n = 24), at 6400 m (n = 14), and at 7950 m (n = 5). Remarkable elevations (200%) in flow in the MCA occurred at 7950 m following removal of breathing supplementary oxygen and breathing air for 20 min. Dilation (*24%) of the MCA occurred at 6400 m, which was further increased to 90% at 7950 m. Oxygen supplementation at this highest altitude rapidly reversed the observed MCA vessel dilation (denoted by blue triangle). Elevations in CBF via cerebral vasodilation were adequate to maintain oxygen delivery, even at these extreme altitudes. Modified from Wilson et al. (2011).

Summary of the major factors acting to increase ( plus) and decrease (minus) CBF during exposure to hypoxia

Summary of the major factors acting to increase ( plus) and decrease (minus) CBF during exposure to hypoxia

Summary of the major factors acting to increase ( plus) and decrease (minus) CBF during exposure to hypoxia. Cao2, arterial oxygen content; CBV, cerebral blood volume; EDHF, endothelium-derived hyperpolarizing factor; ET-1, endothelin-1; HCT, hematocrit; NO, nitric oxide; O2-, superoxide; PGE, prostaglandins; SNA, sympathetic nerve activity; VAH, ventilatory acclimatization to hypoxia/altitude. Modified from Ainslie and Ogoh (2010); Ainslie et al. (2014).

It is clear that many aspects of CBF regulation and brain function at high altitude warrant further investigation. Indeed, several questions remain. For example, over the period of ventilatory acclimatization (weeks to months), how do interactions between the hypoxic ventilatory response, hypercapnic ventilatoy response, hypoxic cerebral vasodilatation, and hypocapnic cerebral vasoconstriction interact to alter CBF? Furthermore, what is the role of NO and/or adenosine in mediating cerebral vasodilation at high altitude? And last, what is the time-course of recovery in CBF following descent to sea level?

 

Cognitive Impairments at High Altitudes and Adaptation

Xiaodan Yan
High Alt Med Biol. 15:141–145, 2014
http://dx.doi.org:/10.1089/ham.2014.1009

High altitude hypoxia has been shown to have significant impact on cognitive performance. This article reviews the aspects in which, and the conditions under which, decreased cognitive performance has been observed at high altitudes. Neural changes related to high altitude hypoxia are also reviewed with respect to their possible contributions to cognitive impairments. In addition, potential adaptation mechanisms are reviewed among indigenous high altitude residents and long-term immigrant residents, with discussions about methodological concerns related to these studies.

The amount of cognitive impairments at high altitudes is related to the chronicity of exposure. Acute exposure usually refers to a duration of several weeks, whereas chronic exposure usually refer to ‘‘extended permanence’’ in the high altitude environment (Virue´s-Ortega and others, 2004). The altitude of ascending or residence is another factor affecting the severity of impairments. This review will first summarize the cognitive impairments in acute exposure, then talk about impairments in chronic exposure, with discussions about the effect of altitudes in corresponding sections.

 

High altitude-related neurocognitive impairments with ascending altitudes

High altitude-related neurocognitive impairments with ascending altitudes

 

 

High altitude-related neurocognitive impairments with ascending altitudes in acute high altitude exposure (Wilson and others, 2009).

human brain consumes about 20% of the total oxygen intake

human brain consumes about 20% of the total oxygen intake

The human brain consumes about 20% of the total oxygen intake, which is disproportional to its size (about 2% of the total body weight). In this figure, oxygen consumption is reflected from glucose consumption in positron emission tomography (PET) (Alavi and Reivich, 2002).

The possibility of adaptation to high altitude hypoxia has always been an intriguing issue. In the acute cases, the human body does have some capacity for acclimatization, which varies significantly for different individuals. The question is, in chronic cases, for example, does growing up at high altitude regions guarantee sufficient adaption to occur to compensate for the risk of cognitive impairments? Existing research tends to suggest that, although some level of adaptation does occur, neural and cognitive impairments are still observed in these populations who are native or long-term residents at high altitude.

Although multiple studies have suggested that growing up at high altitudes is associated with cognitive impairments, it is not to say that adaptation does not happen with prolonged chronic exposure to high altitudes. One study has revealed that as a function of the length of low altitude residence (across the range of 1–5 years), some neuroimaging parameters of original highlanders who grew up at high altitude regions had shown the trend of converging towards the patterns of original low altitude residents, although such changes were not accompanied by statistically significant changes in cognitive performance (Yan and others, 2010). It is possible that, given sufficiently long time for normoxia adaptation, the neural and cognitive impairments associated with high altitude hypoxia may be alleviated to a certain extent.

In summary, various cognitive impairments associated with high altitude hypoxia have been reported from existing studies, which are accompanied by findings about neural impairments, suggesting that these cognitive impairments have legitimate neural basis. The specific relationships between physiological symptoms and cognitive impairments appear to be complicated and require further elucidation. There are cognitive impairments associated with both acute and chronic exposure to high altitudes; however, particular caution should be taken when interpreting the findings about cognitive impairments among native high altitude residents because of the differences
in cultural and socioeconomic factors. Existing studies have suggested that there can be some level of adaptation to high altitudes, in spite of the fact that some neuronal impairment may be irreversible.

Exercise Capacity and Selected Physiological Factors by Ancestry and Residential Altitude: Cross-Sectional Studies of 9–10-Year-Old Children in Tibet

Bianba, Sveinung Berntsen, Lars Bo Andersen, Hein Stigum, et al.
High Alt Med Biol. 2014; 15:162–169
http://dx.doi.org:/10.1089/ham.2013.1084

Aim: Several physiological compensatory mechanisms have enabled Tibetans to live and work at high altitude, including increased ventilation and pulmonary diffusion capacity, both of which serve to increase oxygen transport in the blood. The aim of the present study was to compare exercise capacity (maximal power output) and selected physiological factors (arterial oxygen saturation and heart rate at rest and during maximal exercise, resting hemoglobin concentration, and forced vital capacity) in groups of native Tibetan children living at different residential altitudes (3700 vs. 4300 m above sea level) and across ancestry (native Tibetan vs. Han Chinese children living at the same altitude of 3700 m). Methods: A total of 430 9–10-year-old native Tibetan children from Tingri (4300 m) and 406 native Tibetan and 406 Han Chinese immigrants (77% lowland-born and 33% highland-born) from Lhasa (3700 m) participated in two cross-sectional studies. The maximal power output (Wmax) was assessed using an ergometer cycle. Results: Lhasa Tibetan children had a 20% higher maximal power output (watts/kg) than Tingri Tibetan and 4% higher than Lhasa Han Chinese. Maximal heart rate, arterial oxygen saturation at rest, lung volume, and arterial oxygen saturation were significantly associated with exercise capacity at a given altitude, but could not fully account for the differences in exercise capacity observed between ancestry groups or altitudes. Conclusions: The superior exercise capacity in native Tibetans vs. Han Chinese may reflect a better adaptation to life at high altitude. Tibetans at the lower residential altitude of 3700 m demonstrated a better exercise capacity than residents at a higher altitude of 4300m when measured at their respective residential altitudes. Such altitude- or ancestry-related difference could not be fully attributed to the physiological factors measured.

Group size effects on foraging and vigilance in migratory Tibetan antelope

Xinming Lian, Tongzuo Zhang, Yifan Cao, Jianping Su, Simon Thirgood
Behavioural Processes 76 (2007) 192–197
http://dx.doi.org:/10.1016/j.beproc.2007.05.001

Large group sizes have been hypothesized to decrease predation risk and increase food competition. We investigated group size effects on vigilance and foraging behavior during the migratory period in female Tibetan antelope Pantholops hodgsoni, in the Kekexili Nature Reserve of Qinghai Province, China. During June to August, adult female antelope and yearling females gather in large migratory groups and cross the Qinghai–Tibet highway to calving grounds within the Nature Reserve and return to Qumalai county after calving. Large groups of antelope aggregate in the migratory corridor where they compete for limited food resources and attract the attention of mammalian and avian predators and scavengers. We restricted our sampling to groups of less than 30 antelopes and thus limit our inference accordingly. Focal-animal sampling was used to record the behavior of the free-ranging antelope except for those with lambs. Tibetan antelope spent more time foraging in larger groups but frequency of foraging bouts was not affected by group size. Conversely, the time spent vigilant and frequency of vigilance bouts decreased with increased group size. We suggest that these results are best explained by competition for food and risk of predation.

High altitude exposure alters gene expression levels of DNA repair enzymes, and modulates fatty acid metabolism by SIRT4 induction in human skeletal muscle

Zoltan Acsa, Zoltan Boria, Masaki Takedaa, Peter Osvatha, et al.
Respiratory Physiology & Neurobiology 196 (2014) 33–37
http://dx.doi.org/10.1016/j.resp.2014.02.006

We hypothesized that high altitude exposure and physical activity associated with the attack to Mt Everest could alter mRNA levels of DNA repair and metabolic enzymes and cause oxidative stress-related challenges in human skeletal muscle. Therefore, we have tested eight male mountaineers (25–40 years old) before and after five weeks of exposure to high altitude, which included attacks to peaks above 8000 m. Data gained from biopsy samples from vastus lateralis revealed increased mRNA levels of both cytosolic and mitochondrial superoxide dismutase. On the other hand 8-oxoguanine DNA glycosylase(OGG1) mRNA levels tended to decrease while Ku70 mRNA levels and SIRT6 decreased with altitude exposure. The levels of SIRT1 and SIRT3 mRNA did not change significantly. But SIRT4 mRNA level increased significantly, which could indicate decreases in fatty acid metabolism, since SIRT4 is one of the important regulators of this process. Within the limitations of this human study, data suggest that combined effects of high altitude exposure and physical activity climbing to Mt. Everest, could jeopardize the integrity of the particular chromosome.

High-altitude adaptations in vertebrate hemoglobins

Roy E. Weber
Respiratory Physiology & Neurobiology 158 (2007) 132–142
http://dx.doi.org:/10.1016/j.resp.2007.05.001

Vertebrates at high altitude are subjected to hypoxic conditions that challenge aerobic metabolism. O2 transport from the respiratory surfaces to tissues requires matching between theO2 loading and unloading tensions and theO2-affinity of blood, which is an integrated function of hemoglobin’s intrinsic O2-affinity and its allosteric interaction with cellular effectors (organic phosphates, protons and chloride). Whereas short-term altitudinal adaptations predominantly involve adjustments in allosteric interactions, long-term, genetically-coded adaptations typically involve changes in the structure of the hemoglobin molecules. The latter commonly comprise substitutions of amino acid residues at the effector binding sites, the heme protein contacts, or at inter-subunit contacts that stabilize either the low-affinity (‘Tense’) or the high-affinity (‘Relaxed’) structures of the molecules. Molecular heterogeneity (multiple iso-Hbs with differentiated oxygenation properties) can further broaden the range of physico-chemical conditions where Hb functions under altitudinal hypoxia. This treatise reviews the molecular and cellular mechanisms that adapt hemoglobin-oxygen affinities in mammals, birds and ectothermic vertebrates at high altitude.

Vertebrate animals display remarkable ability to tolerate high altitudes and cope with the concomitant decreases in O2 tension that potentially constrain aerobic life (Monge and Leon-Velarde, 1991;Weber, 1995; Samaja et al., 2003). Compared to an ambient PO2 of approximately 160 mm Hg at sea level, inspired tension approximates only 95 mm Hg for llamas and frogs from Andean habitats above 4000 m, 45 mm Hg for bar-headed geese that fly across the Himalayas, and 33 mm Hg for Ruppell’s griffon that soars at 11,300 m over Africa’s Ivory Coast. Apart from the distinct adaptations manifest in blood’s O2-transporting properties, tolerance to decreased O2 availability may entail reconfigurations at the organ and cellular levels that include a switch to partial anaerobiosis. Driven by needs to reduce aerobic metabolic rate and maintain functional integrity (Ramirez et al., 2007), these pertain to a core triad of adaptations:

  1. metabolic suppression,
  2. tolerance to metabolite (e.g. lactate) accumulation, and
  3. defenses against increased free radicals associated with return to high O2 tensions (Bickler and Buck, 2007).

The response to oxygen lack comprises two phases

  1. defense, which includes metabolic arrest (a suppression of ATP-demand and ATP-supply) and channel arrest (decreases cell membrane permeability), and
  2. rescue, which commonly involves preferential expression of proteins that are implicated in extending metabolic down-regulation (Hochachka et al., 1996).

These responses vary greatly in different species and different tissues. Thus, although mixed-venous lactate concentrations increase strongly in sea-level as well as high-altitude acclimated pigeons that are exposed to altitude (from 1–2 mM at sea level to 5–7 mM at 9000 m) (Weinstein et al., 1985), and humans performing submaximal work at high altitude show a transient ‘lactate paradox’ (lower peak lactate levels that humans living at sea level (Lundby et al., 2000)), many species do not exhibit altitude-related changes in anaerobic metabolism.

Organismic adaptations to survive and perform physical exercise at extreme altitudinal hypoxia are diverse. In birds the undisputed high-altitude champions, where flapping flight may raise the energy demand 10–20-fold compared to resting levels (Scott et al., 2006), a highly efficient “cross-current” ventilation perfusion arrangement in the lungs may increase arterial O2 tensions above the tensions in expired air (Scheid, 1979) and drastically reduce the difference between inhalant and arterial O2 tensions (to 1 mm Hg in bar-headed geese subjected to simulated altitude of 11580 m) (Black and Tenney, 1980). The Andean frog Telmatobius culeus has a highly ‘oversized’ (folded) and vascularized skin that is ventilated by ‘bobbing’ behavior to support water(=skin) breathing. Manifold organismic adaptations moreover include combinations of increased muscle Mb concentrations (Reynafarje and Morrison, 1962) increased muscle capillarization (manifest in mammals and birds (cf. Monge et al., 1991)) and decreased red cell size (seen in amphibians but not high-altitude reptiles (Ruiz et al., 1989; Ruiz et al., 1993)). Amphibians exhibit an interspecific correlation between erythrocyte count and the degree of vascularization of respiratory surfaces and muscle tissues (Hutchison and Szarski, 1965), that reflect differences in their ability to tolerate altitudinal hypoxia.

A sensitivity analysis of the factors that may limit exercise performance identifies high Hb-O2 affinity, together with high total ventilation and high tissue diffusion capacity as the physiological traits that have greatest adaptive benefit for bird flight at extreme high altitude (Scott and Milsom, 2006). Blood O2 affinity is a combination of the intrinsic O2 affinity of the ‘stripped’ (purified) Hb molecules and the interaction of allosteric effectors (like organic phosphates, protons and chloride ions) that decrease Hb-O2 affinity inside the rbcs (Weber and Fago, 2004). Short-term adaptations in O2 affinity are commonly mediated by changes in erythrocytic effectors such as organic phosphates (2,3-diphosphoglycerate, DPG, in mammals, inositol pentaphosphate, IPP, in birds, ATP in reptiles, and ATP and DPG in amphibians), whereas long-term adaptations (that include interspecific ones that are genetically determined) commonly involve changes in Hb structure (amino acid exchanges) that alter Hb’s intrinsic O2 affinity or its sensitivity to allosteric effectors.

Vertebrate Hbs are tetrameric molecules composed of two α (or α-like) chains and two β (or β-like) chains, which in humans consist of 141 and 146 amino acid residues, respectively. Each subunit exhibits a highly characteristic “globin fold” comprised of seven or eight α-helices (labelled A, B, C, etc.) linked by nonhelical (EF, FG) segments, and N- and C-terminal extensions termed NA and HC, respectively. Individual amino acid residues are identified by their sequential positions in chain or/and the helix; thus α1131(H14)-Ser refers to Serine that is the 131st residue of α1 chain and the 14th of the H. During (de-) oxygenation Hb switches between two major structural states:

  1. the high affinity oxygenated R (relaxed) state that prevails at the respiratory surfaces, and
  2. the low affinity, deoxygenated T (tense) state that occurs predominantly in the tissues and is constrained by additional hydrogen bonds and salt bridges.

The Hbs exhibit cooperative homotropic interactions between the O2 binding heme groups (that cause the S-shaped O2 equilibrium curves and increase O2 loading and unloading for a given change in O2 tension) as well as inhibitory, heterotropic interactions between the hemes and the binding sites of effectors that decrease O2 affinity (increase the half-saturation O2 loading tension, P50) and facilitate O2 unloading.

A comparison of Hbs from different species (cf. Perutz, 1983) reveals that variation in the sensitivities to effectors correlates generally with exchanges of very few of the approximately 287 amino acid residues that comprise each αβ dimer. Thus in adult human Hb (HbA) at physiological pH, the majority of the Bohr effect (pH dependence of Hb-O2 affinity that facilitates O2 release in relatively acid working muscles) results from proton binding at the C-terminal residues of the β-chains (β146-His) (cf. Lukin and Ho, 2004). Correspondingly DPG binds to only four β-chain residues (β1-Val, β2-His, β82-Lys and β143-His), CO2 binding (carbamate formation) occurs at the uncharged amino-termini of both chains (α1-Val and β1-Val), and monovalent anions like chloride are considered to bind at one α-chain site (between α1-Val and α131–Ser) and one β-chain site (between  β82-Lys and β1-Val) (cf. Riggs, 1988).

The small number of sites that primarily determine Hb-O2 affinity and its sensitivity to effectors aligns with the neutral theory of molecular evolution (Kimura, 1979), which holds that the majority of amino acid substitutions are non-adaptive and harmless—and facilitates identification of key molecular mechanisms implicated in adaptations at altitude.

The role of effectors in altitude adaptation is aptly illustrated in humans where Hb structure (intrinsic O2 affinity) remains unchanged. Newcomers and permanent residents at moderate altitude (e.g. 2000 m) show increased DPG levels, resulting in a decreased O2 affinity that positions arterial and mixed venous O2 tensions on the steep part of the O2 equilibrium curve, increasing O2 capacitance ([1]bO2) and O2 transport, without materially compromising O2 loading (Turek et al., 1973; Mairbaurl, 1994). The increased DPG correlates with erythropoietin-mediated formation of new rbcs that have higher glycolytic rates and higher DPG and ATP levels than old rbcs. However, faster increases in P50 than in DPG level indicate contributions from other factors, such as chloride and ATP, and Mg ions that neutralize the anionic effectors (Mairbaurl et al., 1993). At higher altitudes (4559 m) increased hyperventilation that drives off CO2 causes respiratory alkalosis (Mairbaurl, 1994). The higher pH increases O2 affinity via the Bohr effect and, offsetting the effect of increased DPG, leads to a similar O2 affinity and arterio-venous O2 saturation  difference as at sea level (Fig.). O2 unloading in the tissues is moreover enhanced by metabolic acidification of capillary blood (Fig.).

Obviously right-shifted curves (that favor O2 unloading) becomes counterproductive at extreme altitudes where O2 loading becomes compromised, predicting that decreased O2 affinity becomes maladaptive under severe hypoxic stress. This is consistent with the observation that a carbamylation-induced increase in blood O2 affinity of rats (that lowers P50 from 27 to 15 mm Hg), increases survival under hypobaric hypoxia equivalent to 9200 meters’ altitude (Eaton et al., 1974). The altitude limit where increased affinity rather than a decreased affinity optimizes tissue O2 supply < 5000 m in man (Samaja et al., 2003)] depends on organismic adaptations (e.g. efficiency of gas exchange) and thus will vary between species. Mammals that permanently inhabit high altitudes and show high blood O2 affinities include the Andean rodent Chinchilla brevicaudata living at 3000–5000 m (blood P50 = 23 mm Hg compared to 38 mm Hg in the rat) (Ostojic et al., 2002). The deer mouse, Peromyscus maniculatus that occurs continuously from sea level to altitudes above 4300 m shows a strong correlation between blood O2 affinity and native altitude (Snyder et al., 1988). That genetically based differences in cofactor levels may contribute to this relationship follows from lower DPG/Hb ratios found in specimens resident, and native to, high altitude than in those from low altitude, after long-term acclimation of both groups to low altitude (Snyder, 1982).

O2 equilibrium curves of human blood illustrating the effects of increases in red cell DPG and pH at high-altitude

O2 equilibrium curves of human blood illustrating the effects of increases in red cell DPG and pH at high-altitude

 

O2 equilibrium curves of human blood illustrating the effects of increases in red cell DPG and pH at high-altitude (4559 m). Solid curves refer to arterial blood (P50 = 26  mm,upper section) and cubical venous blood (P50 = 27.5 mm Hg, lower section); their displacement reflects the Bohr effect. The broken curves depict effects of increased DPG levels (↑DPG) at unchanged pH, increased pH (↑pH) at unchanged DPG, and of decreased tissue pH (↓pH) resulting from higher degrees of metabolic acidification in the tissues. Open and shaded vertical columns indicate O2 unloaded at sea level and 4559 m, respectively, for venous O2 tensions (PvO2) of 25 and 15 mm Hg,respectively [Modified after (Mairbaurl, 1994)].

Camelids. The high blood-O2 affinities in Andean camelids (llama, vicunia, alpaca and guanaco) whose natural habitats exceed 3000 m (Bartels et al., 1963) compared to those of similarly-sized lowland mammals are well-established. In the camelids a β2His→Asn substitution deletes two of the seven DPG contacts in the tetrameric Hb, which increases blood O2 affinity by reducing the DPG effect. Although the intrinsic Hb-O2 affinity is lower in llama than in the related, lowland camel (Bauer et al., 1980), llama blood has a higher O2 affinity due to a three-fold lower DPG-binding than in camel Hb that has the same DPG binding sites as humans (Bauer et al., 1980). In vicunia, a higher O2 affinity than in llama (that has identical β-chains), correlates with the α130Ala→Thr substitution, which introduces a hydroxyl polar group that predictably reduces the chloride binding at adjacent α131Asn residue .

Sheep and goats commonly express two isoforms, HbA and HbB. The heterogeneity is controlled by two autosomal alleles with codominant expression. Whereas individuals expressing HbA have higher blood-O2 affinity than those that express HbB, heterozygotes that express both forms at equimolar concentrations in the same erythrocytes show intermediate affinity. Anemic blood loss induces switching from HbA to HbC that has a similarly high affinity. Hbs A, B and C have identical α-chains but different β[1]-chains. It appears unknown whether altitudinal exposure (which like anemia, induces tissue hypoxia) modulates Hb heterogeneity via selective expression of specific β-chains.

Compared to most mammals that possess one major adult and one major fetal Hb, yak, Poephagus (=Bos) grunniens, a native to altitudes of 3000–6000 m in Tibet, Nepal and Bhutan, has two or four major adult Hbs and two major fetal Hbs. These Hbs exhibit higher intrinsic affinities than closely-related bovine Hb, marked DPG sensitivities and, exceptional amongst mammals, differentiated O2 affinities that indicates an extended range of ambient O2 tensions (and altitudes) in which the composite Hb functions.

(Not shown).  Representation of interchain contacts considered to underly differentiated O2 affinities in Rueppell’s griffon isoHbs A, A , D and D that have identical β- chains but different α- chains. Accordingly the van der Waal’s contact between β134Ile and β1125-Asp in Hbs A , D and D stabilizes the low-affinity, T-state less strongly than the H-bond between Thr 134 and β1125-Asp and thus increases O2 affinity in Hbs A, D and D. Analogously, the hydrogen bonds between α138-β297/99 that stabilize the high-affinity oxystructure (raising O2 affinity in isoHbs D and D) cannot form in HbA and HbA that have Pro at α138.

Ostriches, the largest extant birds, exhibit a β2His→Gln exchange (that reduces phosphate interaction). They moreover ‘use’ ITP (inositol phosphate) that carries fewer negative charges, and predictably has lesser allosteric effect, than IPP (Isaacks et al., 1977), predicting a high blood O2 affinity that is compatible with ‘scaling’ and (as in elephants) increases high altitude tolerance.

Whereas some adult birds express one major iso-Hb (HbA), the majority of species, reportedly all that fly at high altitudes (Hiebl et al., 1987), also express a less abundant HbD. HbD has the same β-chains as HbA but different α-chains (αD) and exhibits higher O2 affinities (Huisman et al., 1964). There is no consistent evidence for hypoxia-induced changes in HbD expression.

An example of how “molecular anatomy is just as key to understanding molecular adaptation as phylogeny and physiological ecology” (Golding and Dean, 1998) is Hb of the high-altitude tolerant bar-headed goose that has a sharply higher blood O2 affinity than that of the closely related graylag goose that is restricted to lower altitudes (P50 = 29.7 and 39.5mmHg at 37 ◦C and pH 7.4). The Hbs differ by only four (greylag→bar-headed) amino acid exchanges: α18Gly→Ser, α63Ala→Val, β125Glu→Asp and α119Pro→Ala. The last mentioned exchange that is unique in birds, predictably increases O2 affinity, by deleting a contact between α1119 and β155 that destabilizes the T-structure (Perutz, 1983). Moreover, Andean ‘goose’ Hb that also has high blood O2 affinity shows β55 Leu→Ser that deletes the same contact. Significantly, two human Hb mutants (α119Pro–Ala and β155Met→Ser) engineered by site-directed mutagenesis to mimic the mutations found in bar-headed and Andean geese possess markedly higher O2 affinities than native HbA.

Although “the study of molecular adaptation has long been fraught with difficulties not the least of which is identifying out the hundreds of amino acid replacements, those few directly responsible for major adaptations” Hb’s adaptations to high altitude are a prime example of how “an amino acid replacement of modest effect at the molecular level causes a dramatic expansion in an ecological niche” [quotations from (Golding et al., 1998)].

However, the pathway of molecular O2 from the respiratory medium to the cellular combustion sites via the Hb molecules is regulated by a symphony of supplementary adaptations that span different levels of biological organization, each of which (according to the principle of symmorphosis) may become maximally recruited in extreme cases (as in birds actively flying above 10,000 m). Apart from hyperventilation, that appears to occur ubiquitously (and increases blood O2 affinity via increased pH), different species subjected to less extreme hypoxic stress utilize different adaptations among the arsenal of organismic, cellular and molecular strategies that favor efficient aerobic utilization of the scarce O2 available at high altitude. No clear correlations exist between the adaptive strategies recruited by different animals on the one hand, and their phylogenetic position, mode of life or ecological niches on the other. An overall limitation is that short-term adaptive adjustments in O2 affinity (that may occur within individual animals) necessarily involves rapid adaptive responses, such as changes in the levels of erythrocytic effectors, whereas the long-term acclimations that have accumulated in permanent high-altitude dwellers during evolutionary development.

Genetic Diversity of Microsatellite DNA Loci of Tibetan Antelope (Chiru, Pantholops hodgsonii) in Hoh Xil National Nature Reserve, Qinghai, China

Hui Zhou, Diqiang Li, Yuguang Zhang, Tao Yang, Yi Liu
J Genetics and Genomics (Formerly Acta Genetica Sinica) 2007; 34(7): 600-607

The Tibetan antelope (Pantholops hodgsonii), indigenous to China, became an endangered species because of considerable reduction both in number and distribution during the 20th century. Presently, it is listed as an AppendixⅠspecies by CITES and as CategoryⅠ by the Key Protected Wildlife List of China. Understanding the genetic diversity and population structure of the Tibetan antelope is significant for the development of effective conservation plans that will ensure the recovery and future persistence of this species. Twenty-five microsatellites were selected to obtain loci with sufficient levels of polymorphism that can provide in-formation for the analysis of population structure. Among the 25 loci that were examined, nine of them showed high levels of genetic diversity. The nine variable loci (MCM38, MNS64, IOBT395, MCMAI, TGLA68, BM1329, BMS1341, BM3501, and MB066) were used to examine the genetic diversity of the Tibetan antelope (n = 75) in Hoh Xil National Nature Reserve(HXNNR), Qinghai, China. The results obtained by estimating the number of population suggested that all the 75 Tibetan antelope samples were from the same population. The mean number of alleles per locus was 9.4 ± 0.5300 (range, 7–12) and the mean effective number of alleles was 6.519 ± 0.5271 (range, 4.676–9.169). The observed mean and expected heterozygosity were 0.844 ± 0.0133 (range, 0.791–0.897) and 0.838 ± 0.0132 (range, 0.786–0.891), respectively. Mean Polymorphism Information Content (PIC) was 0.818 ± 0.0158 (range, 0.753–0.881). The value of Fixation index (Fis) ranged from −0.269 to −0.097 with the mean of −0.163 ± 0.0197. Mean Shannon’s information index was 1.990 ± 0.0719 among nine loci (range, 1.660–2.315). These results provide baseline data for the evaluation of the level of genetic variation in Tibetan antelope, which will be important for the development of conservation strategies in future.

Expression profiling of abundant genes in pulmonary and cardiac muscle tissues of Tibetan Antelope (Pantholops hodgsonii)

Xiaomei Tong, Yingzhong Yang, Weiwei Wang, Zenzhong Bai, et al.
Gene 523 (2013) 187–191
http://dx.doi.org/10.1016/j.gene.2013.03.011

The Tibetan Antelope (TA), which has lived at high altitude for millions of years, was selected as the model species of high hypoxia-tolerant adaptation. Here we constructed two cDNA libraries from lung and cardiac muscle tissues, obtained EST sequences from the libraries, and acquired extensive expression data related energy metabolism genes. Comparative analyses of synonymous (Ks) and nonsynonymous (Ka) substitution rates of nucleus-encoded mitochondrial unigenes among different species revealed that many antelope genes have undergone rapid evolution. Surfactant-associated protein A (SP-A) and surfactant-associated protein B (SP-B) genes in the AT lineage experienced accelerated evolution compared to goat and sheep, and these two genes are highly expressed in the lung tissue. This study suggests that many specific genes of lung and cardiac muscle tissues showed unique expression profiles and may undergo fast adaptive evolution in TA. These data provide useful information for studying on molecular adaptation to high-altitude in humans as well as other mammals.

Exogenous Sphingosine-1-Phosphate Boosts Acclimatization in Rats Exposed to Acute Hypobaric Hypoxia: Assessment of Haematological and Metabolic Effects

Sonam Chawla, Babita Rahar, Mrinalini Singh, Anju Bansal, et al.
PLoS ONE 9(6): e98025. http://dx.doi.org:/10.1371/journal.pone.0098025

Background: The physiological challenges posed by hypobaric hypoxia warrant exploration of pharmacological entities to improve acclimatization to hypoxia. The present study investigates the preclinical efficacy of sphingosine-1-phosphate (S1P) to improve acclimatization to simulated hypobaric hypoxia. Experimental Approach: Efficacy of intravenously administered S1P in improving hematological and metabolic acclimatization was evaluated in rats exposed to simulated acute hypobaric hypoxia (7620 m for 6 hours) following S1P pre-treatment for three days. Major Findings: Altitude exposure of the control rats caused systemic hypoxia, hypocapnia (plausible sign of hyperventilation) and respiratory alkalosis due to suboptimal renal compensation indicated by an overt alkaline pH of the mixed venous blood. This was associated with pronounced energy deficit in the hepatic tissue along with systemic oxidative stress and inflammation. S1P pre-treatment improved blood oxygen-carrying-capacity by increasing hemoglobin, hematocrit, and RBC count, probably as an outcome of hypoxia inducible factor-1a mediated  erythropoiesis and renal S1P receptor 1 mediated hemoconcentation. The improved partial pressure of oxygen in the blood could further restore aerobic respiration and increase ATP content in the hepatic tissue of S1P treated animals. S1P could also protect the animals from hypoxia mediated oxidative stress and inflammation. Conclusion: The study findings highlight S1P’s merits as a preconditioning agent for improving acclimatization to acute hypobaric hypoxia exposure. The results may have long term clinical application for improving physiological acclimatization of subjects venturing into high altitude for occupational or recreational purposes.

S1P Stabilizes HIF-1a and Boosts HIF-1a Mediated Hypoxia Adaptive Responses

S1P pre-conditioning led to 1.9 fold higher HIF-1a level in the kidney tissue (p<0.001) and 1.3 fold higher HIF-1a level in the liver tissue (p<0.001) in 1 mg/kg b.w. S1P group than in hypoxia control group. However, the hypoxia control group also had 1.3 folds higher HIF-1a levels in both liver and kidney tissues than in normoxia control groups, indicating a non-hypoxic boost of HIF-1a in S1P treated animals (Figure 1a and b). Further, plasma Epo levels were also observed to be significantly higher following S1P pre-treatment compared to the hypoxia control groups (p=0.05) (Figure 1a). Epo being primarily secreted by the kidneys and its expression being under regulation of HIF-1a, the raised plasma Epo level could be attributed to higher HIF-1a level in the kidney.

Figure 1. (not shown) Effect of S1P treatment on HIF-1a accumulation and downstream gene expression. a) Renal HIF-1a accumulation and Epo accumulation in plasma. HIF-1a accumulation in the renal tissue homogenate and build-up of erythropoietin in plasma was quantified. b) Hepatic HIF-1a accumulation. c) Effect S1P pre-treatment on circulatory VEGF. Vascular endothelial growth factor (VEGF) was quantified in plasma of experimental animals. These estimations were carried out using sandwich ELISA, and were carried out in triplicates for each experimental animal. Values are representative of mean 6 SD (n = 6). Statistical significance was calculated using ANOVA/post hoc Bonferroni. NC: Normoxia control, HC: Hypoxia control, 1: 1 mg S1P/kg b.w., 10: 10 mg S1P/kg b.w., 100: 100 mg S1P/kg b.w.,  p<0.05 compared with the normoxic control, p<0.01 compared with the normoxic control, p<0.001 compared with the normoxic control,  p<0.05 compared with the hypoxic control,  p<0.01 compared with the hypoxic control,  p<0.001 compared with the hypoxic control. http://dx.doi.org:/10.1371/journal.pone.0098025.g001

Figure 2.(not shown)  Effect of S1P treatment on S1P1 expression in renal tissue. Representative immune-blot of S1P1. Densitometric analysis of blot normalized against the loading control (α-tubulin). Values are representative of mean 6 SD (n = 6). Statistical significance was calculated using ANOVA/post hoc Bonferroni. NC: Normoxia control, HC: Hypoxia control, 1: 1 mg S1P/kg b.w., 10: 10 mg S1P/kg b.w., 100: 100 mg S1P/kg b.w.,  p<0.05 compared with the normoxic control,  p<0.01 compared with the normoxic control, p<0.001 compared with the normoxic control, p< 0.05 compared with the hypoxic control, p<0.01 compared with the hypoxic control, p<0.001 compared with the hypoxic control. http://dx.doi.org:/10.1371/journal.pone.0098025.g002

Cloning of hypoxia-inducible factor 1α cDNA from a high hypoxia tolerant mammal—plateau pika (Ochotona curzoniae)

T.B. Zhao, H.X. Ning, S.S. Zhu, P. Sun, S.X. Xu, Z.J. Chang, and X.Q. Zhao
Biochemical and Biophysical Research Communications 316 (2004) 565–572
http://dx.doi.org:/10.1016/j.bbrc.2004.02.087

Hypoxia-inducible factor 1 is a transcription factor composed of HIF-1α and HIF-1β. It plays an important role in the signal transduction of cell response to hypoxia. Plateau pika (Ochotona curzoniae) is a high hypoxia-tolerant and cold adaptation species living only at 3000–5000m above sea level on the Qinghai-Tibet Plateau. In this study, HIF-1α cDNA of plateau pika was cloned and its expression in various tissues was studied. The results indicated that plateau pika HIF-1α cDNA was highly identical to those of the human (82%), bovine (89%), mouse (82%), and Norway rat (77%). The deduced amino acid sequence (822 bp) showed 90%, 92%, 86%, and 86% identities with those of the human, bovine, house mouse, and Norway rat, respectively. Northern blot analyses detected two isoforms named pLHIF-1α and pSHIF-1α. The HIF-1α mRNA was highly expressed in the brain and kidney, and much less in the heart, lung, liver, muscle, and spleen, which was quite different from the expression pattern of mouse mRNA. Meanwhile, a new variant of plateau pika HIF-1α mRNA was identified by RT-PCR and characterized. The deduced protein, composed of 536 amino acids, lacks a part of the oxygen-dependent degradation domain (ODD), both transactivation domains (TADs), and the nuclear localization signal motif (NLS). Our results suggest that HIF-1α may play an important role in the pika’s adaptation to hypoxia, especially in brain and kidney, and pika HIF-1α function pattern may be different from that of mouse HIF-1α. Furthermore, for the high ratio of HIF-1α homology among the animals, the HIF-1α gene may be a good phylogenetic performer in recovering the true phylogenetic relationships among taxa.

Comparative Proteomics Analyses of Kobresia pygmaea Adaptation to Environment along an Elevational Gradient on the Central Tibetan Plateau

Xiong Li, Yunqiang Yang, Lan Ma, Xudong Sun, et al.
PLoS ONE 9(6): e98410. http://dx.doi.org:/10.1371/journal.pone.0098410

Variations in elevation limit the growth and distribution of alpine plants because multiple environmental stresses impact plant growth, including sharp temperature shifts, strong ultraviolet radiation exposure, low oxygen content, etc. Alpine plants have developed special strategies to help survive the harsh environments of high mountains, but the internal mechanisms remain undefined. Kobresia pygmaea, the dominant species of alpine meadows, is widely distributed in the Southeastern Tibet Plateau, Tibet Autonomous Region, China. In this study, we mainly used comparative proteomics analyses to investigate the dynamic protein patterns for K. pygmaea located at four different elevations (4600, 4800, 4950 and 5100 m). A total of 58 differentially expressed proteins were successfully detected and functionally characterized. The proteins were divided into various functional categories, including material and energy metabolism, protein synthesis and degradation, redox process, defense response, photosynthesis, and protein kinase. Our study confirmed that increasing levels of antioxidant and heat shock proteins and the accumulation of primary metabolites, such as proline and abscisic acid, conferred K. pygmaea with tolerance to the alpine environment. In addition, the various methods K. pygmaea used to regulate material and energy metabolism played important roles in the development of tolerance to environmental stress. Our results also showed that the way in which K. pygmaea mediated stomatal characteristics and photosynthetic pigments constitutes an enhanced adaptation to alpine environmental stress. According to these findings, we concluded that K. pygmaea adapted to the high-elevation environment on the Tibetan Plateau by aggressively accumulating abiotic stress related metabolites and proteins and by the various life events mediated by proteins. Based on the species flexible physiological and biochemical processes, we surmised that environment change has only a slight impact on K. pygmaea except for possible impacts to populations on vulnerable edges of the species’ range
Altered mitochondrial biogenesis and its fusion gene expression is involved in the high-altitude adaptation of rat lung

Loganathan Chitra, Rathanam Boopathy
Respiratory Physiology & Neurobiology 192 (2014) 74– 84
http://dx.doi.org/10.1016/j.resp.2013.12.007

Intermittent hypobaric hypoxia-induced preconditioning (IHH-PC) of rat favored the adaption of lungs to severe HH conditions, possibly through stabilization of mitochondrial function. This is based on the data generated on regulatory coordination of nuclear DNA-encoded mitochondrial biogenesis; dynamics,and mitochondrial DNA (mtDNA)-encoded oxidative phosphorylation (mt-OXPHOS) genes expression. At16th day after start of IHH-PC (equivalent to 5,000 m, 6 h/d, 2 w of treatment), rats were exposed to severe HH stimulation at 9142 m for 6 h. The IHH-PC significantly counteracted the HH-induced effect of increased lung: water content; tissue damage; and oxidant injury. Further, IHH-PC significantly increased the mitochondrial number, mtDNA content and mt- OXPHOS complex activity in the lung tissues. This observation is due to an increased expression of genes involved in mitochondrial biogenesis (PGC-1α,ERRα, NRF1, NRF2 and TFAM), fusion (Mfn1 and Mfn2) and mt OXPHOS. Thus, the regulatory pathway formed by PGC-1α/ERRα/Mfn2 axes is required for the mitochondrial adaptation provoked by IHH-PC regimen to counteract subsequent HH stress.

Molecular characteristics of Tibetan antelope (Pantholops hodgsonii) mitochondrial DNA control region and phylogenetic inferences with related species

  1. Feng, B. Fan, K. Li, Q.D. Zhang, et al.
    Small Ruminant Research 75 (2008) 236–242
    http://dx.doi.org:/10.1016/j.smallrumres.2007.06.011

Although Tibetan antelope (Pantholops hodgsonii) is a distinctive wild species inhabiting the Tibet-Qinghai Plateau, its taxonomic classification within the Bovidae is still unclear and little molecular information has been reported to date. In this study of Tibetan antelope, the complete control regions of mtDNA were sequenced and compared to those of Tibetan sheep (Ovis aries) and goat (Capra hircus). The length of the control region in Tibetan antelope, sheep and goat is 1067, 1181/1106 and 1121 bp, respectively. A 75-bp repeat sequence was found near the 5’ end of the control region of Tibetan antelope and sheep, the repeat numbers of which were two in Tibetan antelope and three or four in sheep. Three major domain regions, including HVI, HVII and central domain, in Tibetan antelope, sheep and goat were outlined, as well as other less conserved blocks, such as CSB-1, CSB-2, ETAS-1 and ETAS-2. NJ cluster analysis of the three species revealed that Tibetan antelope was more closely related to Tibetan sheep than Tibetan goat. These results were further confirmed by phylogenetic analysis using the partial control region sequences of these and 13 other antelope species. Tibetan antelope is better assigned to the Caprinae rather than the Antilopinae subfamily of the Bovidae.

Read Full Post »

Neonatal Pathophysiology

Neonatal Pathophysiology

Writer and Curator: Larry H. Bernstein, MD, FCAP 

 

Introduction

This curation deals with a large and specialized branch of medicine that grew since the mid 20th century in concert with the developments in genetics and as a result of a growing population, with large urban populations, increasing problems of premature deliveries.  The problems of prematurity grew very preterm to very low birth weight babies with special problems.  While there were nurseries, the need for intensive care nurseries became evident in the 1960s, and the need for perinatal care of pregnant mothers also grew as a result of metabolic problems of the mother, intrauterine positioning of the fetus, and increasing numbers of teen age pregnancies as well as nutritional problems of the mother.  There was also a period when the manufacturers of nutritional products displaced the customary use of breast feeding, which was consequential.  This discussion is quite comprehensive, as it involves a consideration of the heart, the lungs, the brain, and the liver, to a large extent, and also the kidneys and skeletal development.

It is possible to outline, with a proportionate emphasis based on frequency and severity, this as follows:

  1. Genetic and metabolic diseases
  2. Nervous system
  3. Cardiovascular
  4. Pulmonary
  5. Skeletal – bone and muscle
  6. Hematological
  7. Liver
  8. Esophagus, stomach, and intestines
  9. Kidneys
  10. Immune system

Fetal Development

Gestation is the period of time between conception and birth when a baby grows and develops inside the mother’s womb. Because it’s impossible to know exactly when conception occurs, gestational age is measured from the first day of the mother’s last menstrual cycle to the current date. It is measured in weeks. A normal gestation lasts anywhere from 37 to 41 weeks.

Week 5 is the start of the “embryonic period.” This is when all the baby’s major systems and structures develop. The embryo’s cells multiply and start to take on specific functions. This is called differentiation. Blood cells, kidney cells, and nerve cells all develop. The embryo grows rapidly, and the baby’s external features begin to form.

Week 6-9:   Brain forms into five different areas. Some cranial nerves are visible. Eyes and ears begin to form. Tissue grows that will the baby’s spine and other bones. Baby’s heart continues to grow and now beats at a regular rhythm. Blood pumps through the main vessels. Your baby’s brain continues to grow. The lungs start to form. Limbs look like paddles. Essential organs begin to grow.

Weeks 11-18: Limbs extended. Baby makes sucking motion. Movement of limbs. Liver and pancreas produce secretions. Muscle and bones developing.

Week 19-21: Baby can hear. Mom feels baby – and quickening.

http://www.nlm.nih.gov/medlineplus/ency/article/002398.htm

fetal-development

fetal-development

https://polination.files.wordpress.com/2014/02/abortion-new-research-into-fetal-development.jpg

Inherited Metabolic Disorders

The original cause of most genetic metabolic disorders is a gene mutation that occurred many, many generations ago. The gene mutation is passed along through the generations, ensuring its preservation.

Each inherited metabolic disorder is quite rare in the general population. Considered all together, inherited metabolic disorders may affect about 1 in 1,000 to 2,500 newborns. In certain ethnic populations, such as Ashkenazi Jews (Jews of central and eastern European ancestry), the rate of inherited metabolic disorders is higher.

Hundreds of inherited metabolic disorders have been identified, and new ones continue to be discovered. Some of the more common and important genetic metabolic disorders include:

Lysosomal storage disorders : Lysosomes are spaces inside cells that break down waste products of metabolism. Various enzyme deficiencies inside lysosomes can result in buildup of toxic substances, causing metabolic disorders including:

  • Hurler syndrome (abnormal bone structure and developmental delay)
  • Niemann-Pick disease (babies develop liver enlargement, difficulty feeding, and nerve damage)
  • Tay-Sachs disease (progressive weakness in a months-old child, progressing to severe nerve damage; the child usually lives only until age 4 or 5)
  • Gauchers disease and others

Galactosemia: Impaired breakdown of the sugar galactose leads to jaundice, vomiting, and liver enlargement after breast or formula feeding by a newborn.

Maple syrup urine disease: Deficiency of an enzyme called BCKD causes buildup of amino acids in the body. Nerve damage results, and the urine smells like syrup.

Phenylketonuria (PKU): Deficiency of the enzyme PAH results in high levels of phenylalanine in the blood. Mental retardation results if the condition is not recognized.

Glycogen storage diseases: Problems with sugar storage lead to low blood sugar levels, muscle pain, and weakness.

Metal metabolism disorders: Levels of trace metals in the blood are controlled by special proteins. Inherited metabolic disorders can result in protein malfunction and toxic accumulation of metal in the body:

Wilson disease (toxic copper levels accumulate in the liver, brain, and other organs)

Hemochromatosis (the intestines absorb excessive iron, which builds up in the liver, pancreas, joints, and heart, causing damage)

Organic acidemias: methylmalonic acidemia and propionic acidemia.

Urea cycle disorders: ornithine transcarbamylase deficiency and citrullinemia

Hemoglobinopathies – thalassemias, sickle cell disease

Red cell enzyme disorders – glucose-6-phosphate dehydrogenase, pyruvate kinase

This list is by no means complete.

http://www.webmd.com/a-to-z-guides/inherited-metabolic-disorder-types-and-treatments

New variations in the galactose-1-phosphate uridyltransferase (GALT) gene

Clinical and molecular spectra in galactosemic patients from neonatal screening in northeastern Italy: Structural and functional characterization of new variations in the galactose-1-phosphate uridyltransferase (GALT) gene

E Viggiano, A Marabotti, AP Burlina, C Cazzorla, MR D’Apice, et al.
Gene 559 (2015) 112–118
http://dx.doi.org/10.1016/j.gene.2015.01.013
Galactosemia (OMIM 230400) is a rare autosomal recessive inherited disorder caused by deficiency of galactose-1-phosphate uridyltransferase (GALT; OMIM 606999) activity. The incidence of galactosemia is 1 in 30,000–60,000, with a prevalence of 1 in 47,000 in the white population. Neonates with galactosemia can present acute symptoms, such as severe hepatic and renal failure, cataract and sepsis after milk introduction. Dietary restriction of galactose determines the clinical improvement in these patients. However, despite early diagnosis by neonatal screening and dietary treatment, a high percentage of patients develop long-term complications such as cognitive disability, speech problems, neurological and/or movement disorders and, in females, ovarian dysfunction.

With the benefit of early diagnosis by neonatal screening and early therapy, the acute presentation of classical galactosemia can be prevented. The objectives of the current study were to report our experience with a group of galactosemic patients identified through the neonatal screening programs in northeastern Italy during the last 30 years.

No neonatal deaths due to galactosemia complications occurred after the introduction of the neonatal screening program. However, despite the early diagnosis and dietary treatment, the patients with classical galactosemia showed one or more long-term complications.

A total of 18 different variations in the GALT gene were found in the patient cohort: 12 missense, 2 frameshift, 1 nonsense, 1 deletion, 1 silent variation, and 1 intronic. Six (p.R33P, p.G83V, p.P244S, p.L267R, p.L267V, p.E271D) were new variations. The most common variation was p.Q188R (12 alleles, 31.5%), followed by p.K285N (6 alleles, 15.7%) and p.N314D (6 alleles, 15.7%). The other variations comprised 1 or 2 alleles. In the patients carrying a new mutation, the biochemical analysis of GALT activity in erythrocytes showed an activity of < 1%. In silico analysis (SIFT, PolyPhen-2 and the computational analysis on the static protein structure) showed potentially damaging effects of the six new variations on the GALT protein, thus expanding the genetic spectrum of GALT variations in Italy. The study emphasizes the difficulty in establishing a genotype–phenotype correlation in classical galactosemia and underlines the importance of molecular diagnostic testing prior to making any treatment.

Diagnosis and Management of Hereditary Hemochromatosis

Reena J. Salgia, Kimberly Brown
Clin Liver Dis 19 (2015) 187–198
http://dx.doi.org/10.1016/j.cld.2014.09.011

Hereditary hemochromatosis (HH) is a diagnosis most commonly made in patients with elevated iron indices (transferrin saturation and ferritin), and HFE genetic mutation testing showing C282Y homozygosity.

The HFE mutation is believed to result in clinical iron overload through altering hepcidin levels resulting in increased iron absorption.

The most common clinical complications of HH include cirrhosis, diabetes, nonischemic cardiomyopathy, and hepatocellular carcinoma.

Liver biopsy should be performed in patients with HH if the liver enzymes are elevated or serum ferritin is greater than 1000 mg/L. This is useful to determine the degree of iron overload and stage the fibrosis.

Treatment of HH with clinical iron overload involves a combination of phlebotomy and/or chelation therapy. Liver transplantation should be considered for patients with HH-related decompensated cirrhosis.

Health economic evaluation of plasma oxysterol screening in the diagnosis of Niemann–Pick Type C disease among intellectually disabled using discrete event simulation

CDM van Karnebeek, Tima Mohammadi, Nicole Tsaod, Graham Sinclair, et al.
Molecular Genetics and Metabolism 114 (2015) 226–232
http://dx.doi.org/10.1016/j.ymgme.2014.07.004

Background: Recently a less invasive method of screening and diagnosing Niemann–Pick C (NP-C) disease has emerged. This approach involves the use of a metabolic screening test (oxysterol assay) instead of the current practice of clinical assessment of patients suspected of NP-C (review of medical history, family history and clinical examination for the signs and symptoms). Our objective is to compare costs and outcomes of plasma oxysterol screening versus current practice in diagnosis of NP-C disease among intellectually disabled (ID) patients using decision-analytic methods.
Methods: A discrete event simulation model was conducted to follow ID patients through the diagnosis and treatment of NP-C, forecast the costs and effectiveness for a cohort of ID patients and compare the outcomes and costs in two different arms of the model: plasma oxysterol screening and routine diagnosis procedure (anno 2013) over 5 years of follow up. Data from published sources and clinical trials were used in simulation model. Unit costs and quality-adjusted life-years (QALYs) were discounted at a 3% annual rate in the base case analysis. Deterministic and probabilistic sensitivity analyses were conducted.
Results: The outcomes of the base case model showed that using plasma oxysterol screening for diagnosis of NP-C disease among ID patients is a dominant strategy. It would result in lower total cost and would slightly improve patients’ quality of life. The average amount of cost saving was $3642 CAD and the incremental QALYs per each individual ID patient in oxysterol screening arm versus current practice of diagnosis NP-C was 0.0022 QALYs. Results of sensitivity analysis demonstrated robustness of the outcomes over the wide range of changes in model inputs.
Conclusion: Whilst acknowledging the limitations of this study, we conclude that screening ID children and adolescents with oxysterol tests compared to current practice for the diagnosis of NP-C is a dominant strategy with clinical and economic benefits. The less costly, more sensitive and specific oxysterol test has potential to save costs to the healthcare system while improving patients’ quality of life and may be considered as a routine tool in the NP-C diagnosis armamentarium for ID. Further research is needed to elucidate its effectiveness in patients presenting characteristics other than ID in childhood and adolescence.

Neurological and Behavioral Disorders

Estrogen receptor signaling during vertebrate development

Maria Bondesson, Ruixin Hao, Chin-Yo Lin, Cecilia Williams, Jan-Åke Gustafsson
Biochimica et Biophysica Acta 1849 (2015) 142–151
http://dx.doi.org/10.1016/j.bbagrm.2014.06.005

Estrogen receptors are expressed and their cognate ligands produced in all vertebrates, indicative of important and conserved functions. Through evolution estrogen has been involved in controlling reproduction, affectingboth the development of reproductive organs and reproductive behavior. This review broadly describes the synthesis of estrogens and the expression patterns of aromatase and the estrogen receptors, in relation to estrogen functions in the developing fetus and child. We focus on the role of estrogens for the development of reproductive tissues, as well as non-reproductive effects on the developing brain. We collate data from human, rodent, bird and fish studies and highlight common and species-specific effects of estrogen signaling on fetal development. Morphological malformations originating from perturbed estrogen signaling in estrogen receptor and aromatase knockout mice are discussed, as well as the clinical manifestations of rare estrogen receptor alpha and aromatase gene mutations in humans. This article is part of a Special Issue entitled: Nuclear receptors in animal development.

 

Memory function and hippocampal volumes in preterm born very-low-birth-weight (VLBW) young adults

Synne Aanes, Knut Jørgen Bjuland, Jon Skranes, Gro C.C. Løhaugen
NeuroImage 105 (2015) 76–83
http://dx.doi.org/10.1016/j.neuroimage.2014.10.023

The hippocampi are regarded as core structures for learning and memory functions, which is important for daily functioning and educational achievements. Previous studies have linked reduction in hippocampal volume to working memory problems in very low birth weight (VLBW; ≤1500 g) children and reduced general cognitive ability in VLBW adolescents. However, the relationship between memory function and hippocampal volume has not been described in VLBW subjects reaching adulthood. The aim of the study was to investigate memory function and hippocampal volume in VLBW young adults, both in relation to perinatal risk factors and compared to term born controls, and to look for structure–function relationships. Using Wechsler Memory Scale-III and MRI, we included 42 non-disabled VLBW and 61 control individuals at age 19–20 years, and related our findings to perinatal risk factors in the VLBW-group. The VLBW young adults achieved lower scores on several subtests of the Wechsler Memory Scale-III, resulting in lower results in the immediate memory indices (visual and auditory), the working memory index, and in the visual delayed and general memory delayed indices, but not in the auditory delayed and auditory recognition delayed indices. The VLBW group had smaller absolute and relative hippocampal volumes than the controls. In the VLBW group inferior memory function, especially for the working memory index, was related to smaller hippocampal volume, and both correlated with lower birth weight and more days in the neonatal intensive care unit (NICU). Our results may indicate a structural–functional relationship in the VLBW group due to aberrant hippocampal development and functioning after preterm birth.

The relation of infant attachment to attachment and cognitive and behavioural outcomes in early childhood

Yan-hua Ding, Xiu Xua, Zheng-yan Wang, Hui-rong Li, Wei-ping Wang
Early Human Development 90 (2014) 459–464
http://dx.doi.org/10.1016/j.earlhumdev.2014.06.004

Background: In China, research on the relation of mother–infant attachment to children’s development is scarce.
Aims: This study sought to investigate the relation of mother–infant attachment to attachment, cognitive and behavioral development in young children.                                                                                                                            Study design: This study used a longitudinal study design.
Subjects: The subjects included healthy infants (n=160) aged 12 to 18 months.
Outcome measures: Ainsworth’s “Strange Situation Procedure” was used to evaluate mother–infant attachment types. The attachment Q-set (AQS) was used to evaluate the attachment between young children and their mothers. The Bayley scale of infant development-second edition (BSID-II) was used to evaluate cognitive developmental level in early childhood. Achenbach’s child behavior checklist (CBCL) for 2- to 3-year-oldswas used to investigate behavioral problems.
Results: In total, 118 young children (73.8%) completed the follow-up; 89.7% of infants with secure attachment and 85.0% of infants with insecure attachment still demonstrated this type of attachment in early childhood (κ = 0.738, p b 0.05). Infants with insecure attachment collectively exhibited a significantly lower mental development index (MDI) in early childhood than did infants with secure attachment, especially the resistant type. In addition, resistant infants were reported to have greater social withdrawal, sleep problems and aggressive behavior in early childhood.
Conclusion: There is a high consistency in attachment development from infancy to early childhood. Secure mother–infant attachment predicts a better cognitive and behavioral outcome; whereas insecure attachment, especially the resistant attachment, may lead to a lower cognitive level and greater behavioral problems in early childhood.

representations of the HPA axis

representations of the HPA axis

representations of limbic stress-integrative pathways from the prefrontal cortex, amygdala and hippocampus

representations of limbic stress-integrative pathways from the prefrontal cortex, amygdala and hippocampus

Fetal programming of schizophrenia: Select mechanisms

Monojit Debnatha, Ganesan Venkatasubramanian, Michael Berk
Neuroscience and Biobehavioral Reviews 49 (2015) 90–104
http://dx.doi.org/10.1016/j.neubiorev.2014.12.003

Mounting evidence indicates that schizophrenia is associated with adverse intrauterine experiences. An adverse or suboptimal fetal environment can cause irreversible changes in brain that can subsequently exert long-lasting effects through resetting a diverse array of biological systems including endocrine, immune and nervous. It is evident from animal and imaging studies that subtle variations in the intrauterine environment can cause recognizable differences in brain structure and cognitive functions in the offspring. A wide variety of environmental factors may play a role in precipitating the emergent developmental dysregulation and the consequent evolution of psychiatric traits in early adulthood by inducing inflammatory, oxidative and nitrosative stress (IO&NS) pathways, mitochondrial dysfunction, apoptosis, and epigenetic dysregulation. However, the precise mechanisms behind such relationships and the specificity of the risk factors for schizophrenia remain exploratory. Considering the paucity of knowledge on fetal programming of schizophrenia, it is timely to consolidate the recent advances in the field and put forward an integrated overview of the mechanisms associated with fetal origin of schizophrenia.

NMDA receptor dysfunction in autism spectrum disorders

Eun-Jae Lee, Su Yeon Choi and Eunjoon Kim
Current Opinion in Pharmacology 2015, 20:8–13
http://dx.doi.org/10.1016/j.coph.2014.10.007

Autism spectrum disorders (ASDs) represent neurodevelopmental disorders characterized by two core symptoms;

(1)  impaired social interaction and communication, and
(2)  restricted and repetitive behaviors, interests, and activities.

ASDs affect ~ 1% of the population, and are considered to be highly genetic in nature. A large number (~600) of ASD-related genetic variations have been identified (sfari.org), and target gene functions are apparently quite diverse. However, some fall onto common pathways, including synaptic function and chromosome remodeling, suggesting that core mechanisms may exist.

Abnormalities and imbalances in neuronal excitatory and inhibitory synapses have been implicated in diverse neuropsychiatric disorders including autism spectrum disorders (ASDs). Increasing evidence indicates that dysfunction of NMDA receptors (NMDARs) at excitatory synapses is associated with ASDs. In support of this, human ASD-associated genetic variations are found in genes encoding NMDAR subunits. Pharmacological enhancement or suppression of NMDAR function ameliorates ASD symptoms in humans. Animal models of ASD display bidirectional NMDAR dysfunction, and correcting this deficit rescues ASD-like behaviors. These findings suggest that deviation of NMDAR function in either direction contributes to the development of ASDs, and that correcting NMDAR dysfunction has therapeutic potential for ASDs.

Among known synaptic proteins implicated in ASD are metabotropic glutamate receptors (mGluRs). Functional enhancement and suppression of mGluR5 are associated with fragile X syndrome and tuberous sclerosis, respectively, which share autism as a common phenotype. More recently, ionotropic glutamate receptors, namely NMDA receptors (NMDARs) and AMPA receptors (AMPARs), have also been implicated in ASDs. In this review, we will focus on NMDA receptors and summarize evidence supporting the hypothesis that NMDAR dysfunction contributes to ASDs, and, by extension, that correcting NMDAR dysfunction has therapeutic potential for ASDs. ASD-related human NMDAR genetic variants.

Chemokines roles within the hippocampus

Chemokines roles within the hippocampus

IL-1 mediates stress-induced activation of the HPA axis

IL-1 mediates stress-induced activation of the HPA axis

A systemic model of the beneficial role of immune processes in behavioral and neural plasticity

A systemic model of the beneficial role of immune processes in behavioral and neural plasticity

Three Classes of Glutamate Receptors

Three Classes of Glutamate Receptors

Clinical studies on ASDs have identified genetic variants of NMDAR subunit genes. Specifically, de novo mutations have been identified in the GRIN2B gene, encoding the GluN2B subunit. In addition, SNP analyses have linked both GRIN2A (GluN2A subunit) and GRIN2B with ASDs. Because assembled NMDARs contain four subunits, each with distinct properties, ASD-related GRIN2A/ GRIN2B variants likely alter the functional properties of NMDARs and/or NMDAR-dependent plasticity.

Pharmacological modulation of NMDAR function can improve ASD symptoms. D-cycloserine (DCS), an NMDAR agonist, significantly ameliorates social withdrawal and repetitive behavior in individuals with ASD. These results suggest that reduced NMDAR function may contribute to the development of ASDs in humans.

We can divide animal studies into two groups. The first group consists of animals in which NMDAR modulators were shown to normalize both NMDAR dysfunction and ASD-like behaviors, establishing strong association between NMDARs and ASD phenotypes (Fig.). In the second group, NMDAR modulators were shown to rescue ASD-like behaviors, but NMDAR dysfunction and its correction have not been demonstrated.

ASD models with data showing rescue of both NMDAR dysfunction and ASD like behaviors Mice lacking neuroligin-1, an excitatory postsynaptic adhesion molecule, show reduced NMDAR function in the hippocampus and striatum, as evidenced by a decrease in NMDA/AMPA ratio and long-term potentiation (LTP). Neuroligin-1 is thought to enhance synaptic NMDAR function, by directly interacting with and promoting synaptic localization of NMDARs.

Fig not shown.

Bidirectional NMDAR dysfunction in animal models of ASD. Animal models of ASD with bidirectional NMDAR dysfunction can be positioned on either side of an NMDAR function curve. Model animals were divided into two groups.

Group 1: NMDAR modulators normalize both NMDAR dysfunction and ASD-like behaviors (green).

Group 2: NMDAR modulators rescue ASD-like behaviors, but NMDAR dysfunction and its rescue have not been demonstrated (orange). Note that Group 2 animals are tentatively placed on the left-hand side of the slope based on the observed DCS rescue of their ASD-like phenotypes, but the directions of their NMDAR dysfunctions remain to be experimentally determined.

ASD models with data showing rescue of ASD-like behaviors but no demonstrated NMDAR dysfunction

Tbr1 is a transcriptional regulator, one of whose targets is the gene encoding the GluN2B subunit of NMDARs. Mice haploinsufficient for Tbr1 (Tbr1+/-) show structural abnormalities in the amygdala and limited GluN2B induction upon behavioral stimulation. Both systemic injection and local amygdalar infusion of DCS rescue social deficits and impaired associative memory in Tbr1+/- mice. However, reduced NMDAR function and its DCS-dependent correction have not been demonstrated.

Spatial working memory and attention skills are predicted by maternal stress during pregnancy

André Plamondon, Emis Akbari, Leslie Atkinson, Meir Steiner
Early Human Development 91 (2015) 23–29
http://dx.doi.org/10.1016/j.earlhumdev.2014.11.004

Introduction: Experimental evidence in rodents shows that maternal stress during pregnancy (MSDP) negatively impacts spatial learning and memory in the offspring. We aim to investigate the association between MSDP (i.e., life events) and spatial working memory, as well as attention skills (attention shifting and attention focusing), in humans. The moderating roles of child sex, maternal anxiety during pregnancy and postnatal care are also investigated.  Methods: Participants were 236mother–child dyads that were followed from the second trimester of pregnancy until 4 years postpartum. Measurements included questionnaires and independent observations.
Results: MSDP was negatively associated with attention shifting at 18monthswhen concurrent maternal anxiety was low. MSDP was associated with poorer spatial working memory at 4 years of age, but only for boys who experienced poorer postnatal care.
Conclusion: Consistent with results observed in rodents, MSDP was found to be associated with spatial working memory and attention skills. These results point to postnatal care and maternal anxiety during pregnancy as potential targets for interventions that aim to buffer children from the detrimental effects of MSDP.

Acute and massive bleeding from placenta previa and infants’ brain damage

Ken Furuta, Shuichi Tokunaga, Seishi Furukawa, Hiroshi Sameshima
Early Human Development 90 (2014) 455–458
http://dx.doi.org/10.1016/j.earlhumdev.2014.06.002

Background: Among the causes of third trimester bleeding, the impact of placenta previa on cerebral palsy is not well known.
Aims: To clarify the effect ofmaternal bleeding fromplacenta previa on cerebral palsy, and in particular when and how it occurs.
Study design: A descriptive study.
Subjects: Sixty infants born to mothers with placenta previa in our regional population-based study of 160,000 deliveries from 1998 to 2012. Premature deliveries occurring atb26 weeks of gestation and placenta accrete were excluded.
Outcome measures: Prevalence of cystic periventricular leukomalacia (PVL) and cerebral palsy (CP).
Results: Five infants had PVL and 4 of these infants developed CP (1/40,000 deliveries). Acute and massive bleeding (>500 g) within 8 h) occurred at around 30–31 weeks of gestation, and was severe enough to deliver the fetus. None of the 5 infants with PVL underwent antenatal corticosteroid treatment, and 1 infant had mild neonatal hypocapnia with a PaCO2 < 25 mm Hg. However, none of the 5 PVL infants showed umbilical arterial academia with pH < 7.2, an abnormal fetal heart rate monitoring pattern, or neonatal hypotension.
Conclusions: Our descriptive study showed that acute and massive bleeding from placenta previa at around 30 weeks of gestation may be a risk factor for CP, and requires careful neonatal follow-up. The underlying process connecting massive placental bleeding and PVL requires further investigation.

Impact of bilirubin-induced neurologic dysfunction on neurodevelopmental outcomes

Courtney J. Wusthoff, Irene M. Loe
Seminars in Fetal & Neonatal Medicine 20 (2015) 52e57
http://dx.doi.org/10.1016/j.siny.2014.12.003

Extreme neonatal hyperbilirubinemia has long been known to cause the clinical syndrome of kernicterus, or chronic bilirubin encephalopathy (CBE). Kernicterus most usually is characterized by choreoathetoid cerebral palsy (CP), impaired upward gaze, and sensorineural hearing loss, whereas cognition is relatively spared. The chronic condition of kernicterus may be, but is not always, preceded in the acute stage by acute bilirubin encephalopathy (ABE). This acute neonatal condition is also due to hyperbilirubinemia, and is characterized by lethargy and abnormal behavior, evolving to frank neonatal encephalopathy, opisthotonus, and seizures. Less completely defined is the syndrome of bilirubin-induced neurologic dysfunction (BIND).

Bilirubin-induced neurologic dysfunction (BIND) is the constellation of neurologic sequelae following milder degrees of neonatal hyperbilirubinemia than are associated with kernicterus. Clinically, BIND may manifest after the neonatal period as developmental delay, cognitive impairment, disordered executive function, and behavioral and psychiatric disorders. However, there is controversy regarding the relative contribution of neonatal hyperbilirubinemia versus other risk factors to the development of later neurodevelopmental disorders in children with BIND. In this review, we focus on the empiric data from the past 25 years regarding neurodevelopmental outcomes and BIND, including specific effects on developmental delay, cognition, speech and language development, executive function, and the neurobehavioral disorders, such as attention deficit/hyperactivity disorder and autism.

As noted in a technical report by the American Academy of Pediatrics Subcommittee on Hyperbilirubinemia, “it is apparent that the use of a single total serum bilirubin level to predict long-term outcomes is inadequate and will lead to conflicting results”. As described above, this has certainly been the case in research to date. To clarify how hyperbilirubinemia influences neurodevelopmental outcome, more sophisticated consideration is needed both of how to assess bilirubin exposure leading to neurotoxicity, and of those comorbid conditions which may lower the threshold for brain injury.

For example, premature infants are known to be especially susceptible to bilirubin neurotoxicity, with kernicterus reported following TB levels far lower than the threshold expected in term neonates. Similarly, among extremely preterm neonates, BBC is proportional to gestational age, meaning that the most premature infants have the highest UB, even for similar TB levels. Thus, future studies must be adequately powered to examine preterm infants separately from term infants, and should consider not just peak TB, but also BBC, as independent variables in neonates with hyperbilirubinemia. Similarly, an analysis by the NICHD NRN found that, among ELBW infants, higher UB levels were associated with a higher risk of death or NDI. However, increased TB levels were only associated with death or NDI in unstable infants. Again, UB or BBC appeared to be more useful than TB.

Are the neuromotor disabilities of bilirubin-induced neurologic dysfunction disorders related to the cerebellum and its connections?

Jon F. Watchko, Michael J. Painter, Ashok Panigrahy
Seminars in Fetal & Neonatal Medicine 20 (2015) 47e51
http://dx.doi.org/10.1016/j.siny.2014.12.004

Investigators have hypothesized a range of subcortical neuropathology in the genesis of bilirubin induced neurologic dysfunction (BIND). The current review builds on this speculation with a specific focus on the cerebellum and its connections in the development of the subtle neuromotor disabilities of BIND. The focus on the cerebellum derives from the following observations:
(i) the cerebellum is vulnerable to bilirubin-induced injury; perhaps the most vulnerable region within the central nervous system;
(ii) infants with cerebellar injury exhibit a neuromotor phenotype similar to BIND; and                                                       (iii) the cerebellum has extensive bidirectional circuitry projections to motor and non-motor regions of the brain-stem and cerebral cortex that impact a variety of neurobehaviors.
Future study using advanced magnetic resonance neuroimaging techniques have the potential to shed new insights into bilirubin’s effect on neural network topology via both structural and functional brain connectivity measurements.

Bilirubin-induced neurologic damage is most often thought of in terms of severe adverse neuromotor (dystonia with or without athetosis) and auditory (hearing impairment or deafness) sequelae. Observed together, they comprise the classic neurodevelopmental phenotype of chronic bilirubin encephalopathy or kernicterus, and may also be seen individually as motor or auditory predominant subtypes. These injuries reflect both a predilection of bilirubin toxicity for neurons (relative to glial cells) and the regional topography of bilirubin-induced neuronal damage characterized by prominent involvement of the globus pallidus, subthalamic nucleus, VIII cranial nerve, and cochlear nucleus.

It is also asserted that bilirubin neurotoxicity may be associated with other less severe neurodevelopmental disabilities, a condition termed “subtle kernicterus” or “bilirubin-induced neurologic dysfunction” (BIND). BIND is defined by a constellation of “subtle neurodevelopmental disabilities without the classical findings of kernicterus that, after careful evaluation and exclusion of other possible etiologies, appear to be due to bilirubin neurotoxicity”. These purportedly include:

(i) mild-to-moderate disorders of movement (e.g., incoordination, clumsiness, gait abnormalities, disturbances in static and dynamic balance, impaired fine motor skills, and ataxia);                                                                                             (ii) disturbances in muscle tone; and
(iii) altered sensorimotor integration. Isolated disturbances of central auditory processing are also included in the spectrum of BIND.

  • Cerebellar vulnerability to bilirubin-induced injury
  • Cerebellar injury phenotypes and BIND
  • Cerebellar projections
Transverse section of cerebellum and brainstem

Transverse section of cerebellum and brainstem

Transverse section of cerebellum and brain-stem from a 34 gestational-week premature kernicteric infant formalin-fixed for two weeks. Yellow staining is evident in the cerebellar dentate nuclei (upper arrow) and vestibular nuclei at the pontomedullary junction (lower arrowhead). Photo is courtesy of Mahmdouha Ahdab-Barmada and reprinted with permission from Taylor-Francis Group (Ahdab Barmada M. The neuropathology of kernicterus: definitions and debate. In: Maisel MJ, Watchko JF editors. Neonatal jaundice. Amsterdam: Harwood Academic Publishers; 2000. p. 75e88

Whether cerebellar injury is primal or an integral part of disturbed neural circuitry in bilirubin-induced CNS damage is unclear. Movement disorders, however, are increasingly recognized to arise from abnormalities of neuronal circuitry rather than localized, circumscribed lesions. The cerebellum has extensive bidirectional circuitry projections to an array of brainstem nuclei and the cerebral cortex that modulate and refine motor activities. In this regard, the cerebellum is characteristically subdivided into three lobes based on neuroanatomic and phylogenetic criteria as well as by their primary afferent and efferent connections. They include:
(i) flocculonodular lobe (archicerebellum);
(ii) anterior lobe (paleocerebellum); and
(iii) posterior lobe (neocerebellum).

The archicerebellum, the oldest division phylogenically, receives extensive input from the vestibular system and is therefore also known as the vestibulocerebellum and is important for equilibrium control. The paleocerebellum, also a primitive region, receives extensive somatosensory input from the spinal cord, including the anterior and posterior spinocerebellar pathways that convey unconscious proprioception, and is therefore also known as the spinocerebellum. The neocerebellum is the most recently evolved region, receives most of the input from the cerebral cortex, and is thus termed the cerebrocerebellum. This area has greatly expanded in association with the extensive development of the cerebral cortex in mammals and especially primates. To cause serious longstanding dysfunction, cerebellar injury must typically involve the deep cerebellar nuclei and their projections.

Schematic of the bidirectional connectivity between the cerebellum and other

Schematic of the bidirectional connectivity between the cerebellum and other

Schematic of the bidirectional connectivity between the cerebellum and other brain regions including the cerebral cortex. Most cerebro-cerebellar afferent projections pass through the basal (anterior or ventral) pontine nuclei and intermediate cerebellar peduncle, whereas most cerebello-cerebral efferent projections pass through the dentate and ventrolateral thalamic nuclei. DCN, deep cerebellar nuclei; RN, red nucleus; ATN, anterior thalamic nucleus; PFC, prefrontal cortex; MC, motor cortex; PC, parietal cortex; TC, temporal cortex; STN, subthalamic nucleus; APN, anterior pontine nuclei. Reprinted under the terms of the Creative Commons Attribution License from D’Angelo E, Casali S. Seeking a unified framework for cerebellar function and dysfunction: from circuit to cognition. Front Neural Circuits 2013; 6:116.

Given the vulnerability of the cerebellum to bilirubin-induced injury, cerebellar involvement should also be evident in classic kernicterus, contributing to neuromotor deficits observed therein. It is of interest, therefore, that cerebellar damage may play a role in the genesis of bilirubin-induced dystonia, a prominent neuromotor feature of chronic bilirubin encephalopathy in preterm and term neonates alike. This complex movement disorder is characterized by involuntary sustained muscle contractions that result in abnormal position and posture. Moreover, dystonia that is brief in duration results in chorea, and, if brief and repetitive, leads to athetosis ‒ conditions also classically observed in kernicterus. Recent evidence suggests that dystonic movements may depend on disruption of both basal ganglia and cerebellar neuronal networks, rather than isolated dysfunction of only one motor system.

Dystonia is also a prominent feature in Gunn rat pups and neonatal Ugt1‒/‒-deficient mice both robust models of kernicterus. The former is used as an experimental model of dystonia. Although these models show basal ganglia injury, the sine qua non of bilirubin-induced murine neuropathology is cerebellar damage and resultant cerebellar hypoplasia.

Studies are needed to define more precisely the motor network abnormalities in kernicterus and BIND. Magnetic resonance imaging (MRI) has been widely used in evaluating infants at risk for bilirubin-induced brain injury using conventional structural T1-and T2-weighted imaging. Infants with chronic bilirubin encephalopathy often demonstrate abnormal bilateral, symmetric, high-signal intensity on T2-weighted MRI of the globus pallidus and subthalamic nucleus, consistent with the neuropathology of kernicterus. Early postnatal MRI of at-risk infants, although frequently showing increased T1-signal in these regions, may give false-positive findings due to the presence of myelin in these structures.

Diffusion tensor imaging and tractography could be used to delineate long-term changes involving specific white matter pathways, further elucidating the neural basis of long-term disability in infants and children with chronic bilirubin encephalopathy and BIND. It will be equally valuable to use blood oxygen level-dependent (BOLD) “resting state” functional MRI to study intrinsic connectivity in order to identify vulnerable brain networks in neonates with kernicterus and BIND. Structural networks of the CNS (connectome) and functional network topology can be characterized in infants with kernicterus and BIND to determine disease-related pattern(s) with respect to both long- and short-range connectivity. These findings have the potential to shed novel insights into the pathogenesis of these disorders and their impact on complex anatomical connections and resultant functional deficits.

Audiologic impairment associated with bilirubin-induced neurologic damage

Cristen Olds, John S. Oghalai
Seminars in Fetal & Neonatal Medicine 20 (2015) 42e46
http://dx.doi.org/10.1016/j.siny.2014.12.006

Hyperbilirubinemia affects up to 84% of term and late preterm infants in the first week of life. The elevation of total serum/plasma bilirubin (TB) levels is generally mild, transitory, and, for most children, inconsequential. However, a subset of infants experiences lifelong neurological sequelae. Although the prevalence of classic kernicterus has fallen steadily in the USA in recent years, the incidence of jaundice in term and premature infants has increased, and kernicterus remains a significant problem in the global arena. Bilirubin-induced neurologic dysfunction (BIND) is a spectrum of neurological injury due to acute or sustained exposure of the central nervous system(CNS) to bilirubin. The BIND spectrum includes kernicterus, acute bilirubin encephalopathy, and isolated neural pathway dysfunction.

Animal studies have shown that unconjugated bilirubin passively diffuses across cell membranes and the blood‒brain barrier (BBB), and bilirubin not removed by organic anion efflux pumps accumulates within the cytoplasm and becomes toxic. Exposure of neurons to bilirubin results in increased oxidative stress and decreased neuronal proliferation and presynaptic neuro-degeneration at central glutaminergic synapses. Furthermore, bilirubin administration results in smaller spiral ganglion cell bodies, with decreased cellular density and selective loss of large cranial nerve VIII myelinated fibers. When exposed to bilirubin, neuronal supporting cells have been found to secrete inflammatory markers, which contribute to increased BBB permeability and bilirubin loading.

The jaundiced Gunn rat is the classic animal model of bilirubin toxicity. It is homozygous for a premature stop codon within the gene for UDP-glucuronosyltransferase family 1 (UGT1). The resultant gene product has reduced bilirubin-conjugating activity, leading to a state of hyperbilirubinemia. Studies with this rat model have led to the concept that impaired calcium homeostasis is an important mechanism of neuronal toxicity, with reduced expression of calcium-binding proteins in affected cells being a sensitive index of bilirubin-induced neurotoxicity. Similarly, application of bilirubin to cultured auditory neurons from brainstem cochlear nuclei results in hyperexcitability and excitotoxicity.

The auditory pathway and normal auditory brainstem response (ABR).

The auditory pathway and normal auditory brainstem response (ABR).

The auditory pathway and normal auditory brain-stem response (ABR). The ipsilateral (green) and contralateral (blue) auditory pathways are shown, with structures that are known to be affected by hyperbilirubinemia highlighted in red. Roman numerals in parentheses indicate corresponding waves in the normal human ABR (inset). Illustration adapted from the “Ear Anatomy” series by Robert Jackler and Christine Gralapp, with permission.

Bilirubin-induced neurologic dysfunction (BIND)

Vinod K. Bhutani, Ronald Wong
Seminars in Fetal & Neonatal Medicine 20 (2015) 1
http://dx.doi.org/10.1016/j.siny.2014.12.010

Beyond the traditional recognized areas of fulminant injury to the globus pallidus as seen in infants with kernicterus, other vulnerable areas include the cerebellum, hippocampus, and subthalamic nuclear bodies as well as certain cranial nerves. The hippocampus is a brain region that is particularly affected by age related morphological changes. It is generally assumed that a loss in hippocampal volume results in functional deficits that contribute to age-related cognitive deficits. Lower grey matter volumes within the limbic-striato-thalamic circuitry are common to other etiological mechanisms of subtle neurologic injury. Lower grey matter volumes in the amygdala, caudate, frontal and medial gyrus are found in schizophrenia and in the putamen in autism. Thus, in terms of brain volumetrics, schizophrenia and autism spectrum disorders have a clear degree of overlap that may reflect shared etiological mechanisms. Overlap with injuries observed in infants with BIND raises the question about how these lesions are arrived at in the context of the impact of common etiologies.

Stress-induced perinatal and transgenerational epigenetic programming of brain development and mental health

Olena Babenko, Igor Kovalchuk, Gerlinde A.S. Metz
Neuroscience and Biobehavioral Reviews 48 (2015) 70–91
http://dx.doi.org/10.1016/j.neubiorev.2014.11.013

Research efforts during the past decades have provided intriguing evidence suggesting that stressful experiences during pregnancy exert long-term consequences on the future mental wellbeing of both the mother and her baby. Recent human epidemiological and animal studies indicate that stressful experiences in utero or during early life may increase the risk of neurological and psychiatric disorders, arguably via altered epigenetic regulation. Epigenetic mechanisms, such as miRNA expression, DNA methylation, and histone modifications are prone to changes in response to stressful experiences and hostile environmental factors. Altered epigenetic regulation may potentially influence fetal endocrine programming and brain development across several generations. Only recently, however, more attention has been paid to possible transgenerational effects of stress. In this review we discuss the evidence of transgenerational epigenetic inheritance of stress exposure in human studies and animal models. We highlight the complex interplay between prenatal stress exposure, associated changes in miRNA expression and DNA methylation in placenta and brain and possible links to greater risks of schizophrenia, attention deficit hyperactivity disorder, autism, anxiety- or depression-related disorders later in life. Based on existing evidence, we propose that prenatal stress, through the generation of epigenetic alterations, becomes one of the most powerful influences on mental health in later life. The consideration of ancestral and prenatal stress effects on lifetime health trajectories is critical for improving strategies that support healthy development and successful aging.

Sensitive time-windows for susceptibility in neurodevelopmental disorders

Rhiannon M. Meredith, Julia Dawitz and Ioannis Kramvis
Trends in Neurosciences, June 2012; 35(6): 335-344
http://dx.doi.org:/10.1016/j.tins.2012.03.005

Many neurodevelopmental disorders (NDDs) are characterized by age-dependent symptom onset and regression, particularly during early postnatal periods of life. The neurobiological mechanisms preceding and underlying these developmental cognitive and behavioral impairments are, however, not clearly understood. Recent evidence using animal models for monogenic NDDs demonstrates the existence of time-regulated windows of neuronal and synaptic impairments. We propose that these developmentally-dependent impairments can be unified into a key concept: namely, time-restricted windows for impaired synaptic phenotypes exist in NDDs, akin to critical periods during normal sensory development in the brain. Existence of sensitive time-windows has significant implications for our understanding of early brain development underlying NDDs and may indicate vulnerable periods when the brain is more susceptible to current therapeutic treatments.

Fig (not shown)

Misregulated mechanisms underlying spine morphology in NDDs. Several proteins implicated in monogenic NDDs (highlighted in red) are linked to the regulation of the synaptic cytoskeleton via F-actin through different Rho-mediated signaling pathways (highlighted in green). Mutations in OPHN1, TSC1/2, FMRP, p21-activated kinase (PAK) are directly linked to human NDDs of intellectual disability. For instance, point mutations in OPHN1 and a PAK isoform are linked to non-syndromic mental retardation, whereas mutations or altered expression of TSC1/2 and FMRP are linked to TSC and FXS, respectively. Cytoplasmic interacting protein (CYFIP) and LIM-domain kinase 1 (LIMK1) are known to interact with FMRP and PAK, respectively [105]. LIMK1 is one of many dysregulated proteins contributing to the NDD Williams syndrome. Mouse models are available for all highlighted (red) proteins and reveal specific synaptic and behavioral deficits. Local protein synthesis in synapses, dendrites and glia is also regulated by proteins such as TSC1/2 and the FMRP/CYFIP complex. Abbreviations: 4EBP, 4E binding protein; eIF4E, eukaryotic translation initiation factor 4E.

Fig (not shown)

Sensitive time-windows, synaptic phenotypes and NDD gene targets. Sensitive time-windows exist in neural circuits, during which gene targets implicated in NDDs are normally expressed. Misregulation of these genes can affect multiple synaptic phenotypes during a restricted developmental period. The effect upon synaptic phenotypes is dependent upon the temporal expression of these NDD genes and their targets. (a) Expression outside a critical period of development will have no effect upon synaptic phenotypes. (b,c) A temporal expression pattern that overlaps with the onset (b) or closure (c) of a known critical period can alter the synaptic phenotype during that developmental time-window.

Outstanding questions

(1) Can treatment at early presymptomatic stages in animal models for NDDs prevent or ease the later synaptic, neuronal, and behavioral impairments?

(2) Are all sensory critical periods equally misregulated in mouse models for a specific NDD? Are there different susceptibilities for auditory, visual and somatosensory neurocircuits that reflect the degree of impairments observed in patients?

(3) If one critical period is missed or delayed during formation of a layer-specific connection in a network, does the network overcome this misregulated connectivity or plasticity window?

(4) In monogenic NDDs, does the severity of misregulating one particular time-window for synaptic establishment during development correlate with the importance of that gene for that synaptic circuit?

(5) Why do critical periods close in brain development?

(6) What underlies the regression of some altered synaptic phenotypes in Fmr1-KO mice?

(7) Can the concept of susceptible time-windows be applied to other NDDs, including schizophrenia and Tourette’s syndrome?

Cardiovascular

Cardiac output monitoring in newborns

Willem-Pieter de Boode
Early Human Development 86 (2010) 143–148
http://dx.doi.org:/10.1016/j.earlhumdev.2010.01.032

There is an increased interest in methods of objective cardiac output measurement in critically ill patients. Several techniques are available for measurement of cardiac output in children, although this remains very complex in newborns. Cardiac output monitoring could provide essential information to guide hemodynamic management. An overview is given of various methods of cardiac output monitoring with advantages and major limitations of each technology together with a short explanation of the basic principles.

Fick principle

According to the Fick principle the volume of blood flow in a given period equals the amount of substance entering the blood stream in the same period divided by the difference in concentrations of the substrate upstream respectively downstream to the point of entry in the circulation. This substance can be oxygen (O2-Fick) or carbon dioxide (CO2-FICK), so cardiac output can be calculated by dividing measured pulmonary oxygen uptake by the arteriovenous oxygen concentration difference. The direct O2-Fick method is regarded as gold standard in cardiac output monitoring in a research setting, despite its limitations. When the Fick principle is applied for carbon dioxide (CO2 Fick), the pulmonary carbon dioxide exchange is divided by the venoarterial CO2 concentration difference to calculate cardiac output.

In the modified CO2 Fick method pulmonary CO2 exchange is measured at the endotracheal tube. Measurement of total CO2 concentration in blood is more complex and simultaneous sampling of arterial and central venous blood is required. However, frequent blood sampling will result in an unacceptable blood loss in the neonatal population.

Blood flow can be calculated if the change in concentration of a known quantity of injected indicator is measured in time distal to the point of injection, so an indicator dilution curve can be obtained. Cardiac output can then be calculated with the use of the Stewart–Hamilton equation. Several indicators are used, such as indocyanine green, Evans blue and brilliant red in dye dilution, cold solutions in thermodilution, lithium in lithium dilution, and isotonic saline in ultrasound dilution.

Cardiovascular adaptation to extra uterine life

Alice Lawford, Robert MR Tulloh
Paediatrics And Child Health 2014; 25(1): 1-6.

The adaptation to extra uterine life is of interest because of its complexity and the ability to cause significant health concerns. In this article we describe the normal changes that occur and the commoner abnormalities that are due to failure of normal development and the effect of congenital cardiac disease. Abnormal development may occur as a result of problems with the mother, or with the fetus before birth. After birth it is essential to determine whether there is an underlying abnormality of the fetal pulmonary or cardiac development and to determine the best course of management of pulmonary hypertension or congenital cardiac disease. Causes of underdevelopment, maldevelopment and maladaptation are described as are the causes of critical congenital heart disease. The methods of diagnosis and management are described to allow the neonatologist to successfully manage such newborns.

Fetal vascular structures that exist to direct blood flow

Fetal structure Function
Arterial duct Connects pulmonary artery to the aorta and shunts blood right to left; diverting flow away from fetal lungs
Foramen ovale Opening between the two atria thatdirects blood flow returning to right

atrium through the septal wall into the left atrium bypassing lungs

Ductus venosus Receives oxygenated blood fromumbilical vein and directs it to the

inferior vena cava and right atrium

Umbilical arteries Carrying deoxygenated blood fromthe fetus to the placenta
Umbilical vein Carrying oxygenated blood from theplacenta to the fetus

Maternal causes of congenital heart disease

Maternal disorders rubella, SLE, diabetes mellitus
Maternal drug use Warfarin, alcohol
Chromosomal abnormality Down, Edward, Patau, Turner, William, Noonan

 

Fetal and Neonatal Circulation  The fetal circulation is specifically adapted to efficiently exchange gases, nutrients, and wastes through placental circulation. Upon birth, the shunts (foramen ovale, ductus arteriosus, and ductus venosus) close and the placental circulation is disrupted, producing the series circulation of blood through the lungs, left atrium, left ventricle, systemic circulation, right heart, and back to the lungs.

Clinical monitoring of systemic hemodynamics in critically ill newborns

Willem-Pieter de Boode
Early Human Development 86 (2010) 137–141
http://dx.doi.org:/10.1016/j.earlhumdev.2010.01.031

Circulatory failure is a major cause of mortality and morbidity in critically ill newborn infants. Since objective measurement of systemic blood flow remains very challenging, neonatal hemodynamics is usually assessed by the interpretation of various clinical and biochemical parameters. An overview is given about the predictive value of the most used indicators of circulatory failure, which are blood pressure, heart rate, urine output, capillary refill time, serum lactate concentration, central–peripheral temperature difference, pH, standard base excess, central venous oxygen saturation and color.

Key guidelines

➢ The clinical assessment of cardiac output by the interpretation of indirect parameters of systemic blood flow is inaccurate, irrespective of the level of experience of the clinician

➢ Using blood pressure to diagnose low systemic blood flow will consequently mean that too many patients will potentially be undertreated or overtreated, both with substantial risk of adverse effects and iatrogenic damage.

➢ Combining different clinical hemodynamic parameters enhances the predictive value in the detection of circulatory failure, although accuracy is still limited.

➢ Variation in time (trend monitoring) might possibly be more informative than individual, static values of clinical and biochemical parameters to evaluate the adequacy of neonatal circulation.

Monitoring oxygen saturation and heart rate in the early neonatal period

J.A. Dawson, C.J. Morley
Seminars in Fetal & Neonatal Medicine 15 (2010) 203e207
http://dx.doi.org:/10.1016/j.siny.2010.03.004

Pulse oximetry is commonly used to assist clinicians in assessment and management of newly born infants in the delivery room (DR). In many DRs, pulse oximetry is now the standard of care for managing high risk infants, enabling immediate and dynamic assessment of oxygenation and heart rate. However, there is little evidence that using pulse oximetry in the DR improves short and long term outcomes. We review the current literature on using pulse oximetry to measure oxygen saturation and heart rate and how to apply current evidence to management in the DR.

Practice points

  • Understand how SpO2 changes in the first minutes after birth.
  • Apply a sensor to an infant’s right wrist as soon as possible after birth.
  • Attach sensor to infant then to oximeter cable.
  • Use two second averaging and maximum sensitivity.

Using pulse oximetry assists clinicians:

  1. Assess changes in HR in real time during transition.
  2. Assess oxygenation and titrate the administration of oxygen to maintain oxygenation within the appropriate range for SpO2 during the first minutes after birth.

Research directions

  • What are the appropriate centiles to target during the minutes after birth to prevent hypoxia and hyperoxia: 25th to 75th, or 10th to 90th, or just the 50th (median)?
  • Can the inspired oxygen be titrated against the SpO2 to keep the SpO2 in the ‘normal range’?
  • Does the use of centile charts in the DR for HR and oxygen saturation reduce the rate of hyperoxia when infants are treated with oxygen.
  • Does the use of pulse oximetry immediately after birth improve short term outcomes, e.g. efficacy of immediate respiratory support, intubation rates in the DR, percentage of inspired oxygen, rate of use of adrenalin or chest compressions, duration of hypoxia/hyperoxia and bradycardia.
  • Does the use of pulse oximetry in the DR improve short term respiratory and long term neurodevelopmental outcomes for preterm infants, e.g. rate of intubation, use of surfactant, and duration of ventilation, continuous positive airway pressure, or supplemental oxygen?
  • Can all modern pulse oximeters be used effectively in the DR or do some have a longer delay before giving an accurate signal and more movement artefact?
  • Would a longer averaging time result in more stable data?

Peripheral haemodynamics in newborns: Best practice guidelines

Michael Weindling, Fauzia Paize
Early Human Development 86 (2010) 159–165
http://dx.doi.org:/10.1016/j.earlhumdev.2010.01.033

Peripheral hemodynamics refers to blood flow, which determines oxygen and nutrient delivery to the tissues. Peripheral blood flow is affected by vascular resistance and blood pressure, which in turn varies with cardiac function. Arterial oxygen content depends on the blood hemoglobin concentration (Hb) and arterial pO2; tissue oxygen delivery depends on the position of the oxygen-dissociation curve, which is determined by temperature and the amount of adult or fetal hemoglobin. Methods available to study tissue perfusion include near-infrared spectroscopy, Doppler flowmetry, orthogonal polarization spectral imaging and the peripheral perfusion index. Cardiac function, blood gases, Hb, and peripheral temperature all affect blood flow and oxygen extraction. Blood pressure appears to be less important. Other factors likely to play a role are the administration of vasoactive medications and ventilation strategies, which affect blood gases and cardiac output by changing the intrathoracic pressure.

graphic

NIRS with partial venous occlusion to measure venous oxygen saturation

NIRS with partial venous occlusion to measure venous oxygen saturation

NIRS with partial venous occlusion to measure venous oxygen saturation. Taken from Yoxall and Weindling

Schematic representation of the biphasic relationship between oxygen delivery and oxygen consumption in tissue

Schematic representation of the biphasic relationship between oxygen delivery and oxygen consumption in tissue

graphic

Schematic representation of the biphasic relationship between oxygen delivery and oxygen consumption in tissue.  (a) oxygen delivery (DO2). (b) As DO2 decreases, VO2 is dependent on DO2. The slope of the line indicates the FOE, which in this case is about 0.50. (c) The slope of the line indicates the FOE in the normal situation where oxygenation is DO2 independent, usually < 0.35

The oxygen-dissociation curve

The oxygen-dissociation curve

graphic

The oxygen-dissociation curve

Considerable information about the response of the peripheral circulation has been obtained using NIRS with venous occlusion. Although these measurements were validated against blood co-oximetry in human adults and infants, they can only be made intermittently by a trained operator and are thus not appropriate for general clinical use. Further research is needed to find other better measures of peripheral perfusion and oxygenation which may be easily and continuously monitored, and which could be useful in a clinical setting.

Peripheral oxygenation and management in the perinatal period

Michael Weindling
Seminars in Fetal & Neonatal Medicine 15 (2010) 208e215
http://dx.doi.org:/10.1016/j.siny.2010.03.005

The mechanisms for the adequate provision of oxygen to the peripheral tissues are complex. They involve control of the microcirculation and peripheral blood flow, the position of the oxygen dissociation curve including the proportion of fetal and adult hemoglobin, blood gases and viscosity. Systemic blood pressure appears to have little effect, at least in the non-shocked state. The adequate delivery of oxygen (DO2) depends on consumption (VO2), which is variable. The balance between VO2 and DO2 is given by fractional oxygen extraction (FOE ¼ VO2/DO2). FOE varies from organ to organ and with levels of activity. Measurements of FOE for the whole body produce a range of about 0.15-0.33, i.e. the body consumes 15-33% of oxygen transported.

Fig (not shown)

Biphasic relationship between oxygen delivery (DO2) and oxygen consumption (VO2) in tissue. Dotted lines show fractional oxygen extraction (FOE). ‘A’ indicates the normal situation when VO2 is independent ofDO2 and FOE is about 0.30. AsDO2 decreases in the direction of the arrow, VO2 remains independent of DO2 until the critical point is reached at ‘B’; in this illustration, FOE is about 0.50. The slope of the dotted line indicates the FOE (¼ VO2/DO2), which increases progressively as DO2 decreases.

Relationship between haemoglobin F fraction (HbF) and peripheral fractional oxygen extraction

Relationship between haemoglobin F fraction (HbF) and peripheral fractional oxygen extraction

Graphic
(A)Relationship between haemoglobin F fraction (HbF) and peripheral fractional oxygen extraction in anaemic and control infants. (From Wardle et al.)  (B) HbF synthesis and concentration. (From Bard and Widness.) (C) Oxygen dissociation curve.

Peripheral fractional oxygen extraction in babies

Peripheral fractional oxygen extraction in babies

graphic

Peripheral fractional oxygen extraction in babies with asymptomatic or symptomatic anemia compared to controls. Bars represent the median for each group. (From Wardle et al.)

Practice points

  • Peripheral tissue DO2 is complex: cardiac function, blood gases, Hb concentration and the proportion of HbF, and peripheral temperature all play a part in determining blood flow and oxygen extraction in the sick, preterm infant. Blood pressure appears to be less important.
  • Other factors likely to play a role are the administration of vasoactive medications and ventilation strategies, which affect blood gases and cardiac output by changing intrathoracic pressure.
  • Central blood pressure is a poor surrogate measurement for the adequacy of DO2 to the periphery. Direct measurement, using NIRS, laser Doppler flowmetry or other means, may give more useful information.
  • Reasons for total hemoglobin concentration (Hb) being a relatively poor indicator of the adequacy of the provision of oxygen to the tissues:
  1. Hb is only indirectly related to red blood cell volume, which may be a better indicator of the body’s oxygen delivering capacity.
  2. Hb-dependent oxygen availability depends on the position of the oxygen-hemoglobin dissociation curve.
  3. An individual’s oxygen requirements vary with time and from organ to organ. This means that DO2 also needs to vary.
  4. It is possible to compensate for a low Hb by increasing cardiac output and ventilation, and so the ability to compensate for anemia depends on an individual’s cardio-respiratory reserve as well as Hb.
  5. The normal decrease of Hb during the first few weeks of life in both full-term and preterm babies usually occurs without symptoms or signs of anemia or clinical consequences.

The relationship between VO2 and DO2 is complex and various factors need to be taken into account, including the position of the oxygen dissociation curve, determined by the proportion of HbA and HbF, temperature and pH. Furthermore, diffusion of oxygen from capillaries to the cell depends on the oxygen tension gradient between erythrocytes and the mitochondria, which depends on microcirculatory conditions, e.g. capillary PO2, distance of the cell from the capillary (characterized by intercapillary distances) and the surface area of open capillaries. The latter can change rapidly, for example, in septic shock where arteriovenous shunting occurs associated with tissue hypoxia in spite of high DO2 and a low FOE.

Changes in local temperature deserve particular consideration. When the blood pressure is low, there may be peripheral vasoconstriction with decreased local perfusion and DO2. However, the fall in local tissue temperature would also be expected to be associated with a decreased metabolic rate and a consequent decrease in VO2. Thus a decreased DO2 may still be appropriate for tissue needs.

Pulmonary

Accurate Measurements of Oxygen Saturation in Neonates: Paired Arterial and Venous Blood Analyses

Shyang-Yun Pamela K. Shiao
Newborn and Infant Nurs Rev,  2005; 5(4): 170–178
http://dx.doi.org:/10.1053/j.nainr.2005.09.001

Oxygen saturation (So2) measurements (functional measurement, So2; and fractional measurement, oxyhemoglobin [Hbo2]) and monitoring are commonly investigated as a method of assessing oxygenation in neonates. Differences exist between the So2 and Hbo2 when blood tests are performed, and clinical monitors indicate So2 values. Oxyhemoglobin will decrease with the increased levels of carbon monoxide hemoglobin (Hbco) and methemo-globin (MetHb), and it is the most accurate measurements of oxygen (O2) association of hemoglobin (Hb). Pulse oximeter (for pulse oximetry saturation [Spo2] measurement) is commonly used in neonates. However, it will not detect the changes of Hb variations in the blood for accurate So2 measurements. Thus, the measurements from clinical oximeters should be used with caution. In neonates, fetal hemoglobin (HbF) accounts for most of the circulating Hb in their blood. Fetal hemoglobin has a high O2 affinity, thus releases less O2 to the body tissues, presenting a left-shifted Hbo2 dissociation curve.5,6 To date, however, limited data are available with HbF correction, for accurate arterial and venous (AV) So2 measurements (arterial oxygen saturation [Sao2] and venous oxygen saturation [Svo2]) in neonates, using paired AV blood samples.

In a study of critically ill adult patients, increased pulmonary CO production and elevation in arterial Hbco but not venous Hbco were documented by inflammatory stimuli inducing pulmonary heme oxygenase–1. In normal adults, venous Hbco level might be slightly higher than or equal to arterial Hbco because of production of CO by enzyme heme oxygenase–2, which is predominantly produced in the liver and spleen. However, hypoxia or pulmonary inflammation could induce heme oxygenase–1 to increase endogenous CO, thus elevating pulmonary arterial and systemic arterial Hbco levels in adults. Both endogenous and exogenous CO can suppress proliferation of pulmonary smooth muscles, a significant consideration for the prevention of chronic lung diseases in newborns. Despite these considerations, a later study in healthy adults indicated that the AV differences in Hbco were from technical artifacts and perhaps from inadequate control of different instruments. Thus, further studies are needed to provide more definitive answers for the AV differences of Hbco for adults and neonates with acute and chronic lung diseases.

Methemoglobin is an indicator of Hb oxidation and is essential for accurate measurement of Hbo2, So2, and oxygenation status. No evidence exists to show the AV MetHb difference, although this difference was elucidated with the potential changes of MetHb with different O2 levels.  Methemoglobin can be increased with nitric oxide (NO) therapy, used in respiratory distress syndrome (RDS) to reduce pulmonary hypertension and during heart surgery. Nitric oxide, in vitro, is an oxidant of Hb, with increased O2 during ischemia reperfusion. In hypoxemic conditions in vivo, nitrohemoglobin is a product generated by vessel responsiveness to nitrovasodilators. Nitro-hemoglobin can be spontaneously reversible in vivo, requiring no chemical agents or reductase. However, when O2 levels were increased experimentally in vitro following acidic conditions (pH 6.5) to simulate reperfusion conditions, MetHb levels were increased for the hemolysates (broken red cells). Nitrite-induced oxidation of Hb was associated with an increase in red blood cell membrane rigidity, thus contributing to Hb breakdown. A newer in vitro study of whole blood cells, however, concluded that MetHb formation is not dependent on increased O2 levels. Additional studies are needed to examine in vivo reperfusion of O2 and MetHb effects.

Purpose: The aim of this study was to examine the accuracy of arterial oxygen saturation (Sao2) and venous oxygen saturation (Svo2) with paired arterial and venous (AV) blood in relation to pulse oximetry saturation (Spo2) and oxyhemoglobin (Hbo2) with fetal hemoglobin determination, and their Hbo2 dissociation curves. Method: Twelve preterm neonates with gestational ages ranging from 27 to 34 weeks at birth, who had umbilical AV lines inserted, were investigated. Analyses were performed with 37 pairs of AV blood samples by using a blood volume safety protocol. Results: The mean differences between Sao2 and Svo2, and AV Hbo2 were both 6 percent (F6.9 and F6.7 percent, respectively), with higher Svo2 than those reported for adults. Biases were 2.1 – 0.49 for Sao2, 2.0 – 0.44 for Svo2, and 3.1 – 0.45 for Spo2, compared against Hbo2. With left-shifted Hbo2 dissociation curves in neonates, for the critical values of oxygen tension values between 50 and 75 millimeters of mercury, Hbo2 ranged from 92 to 93.4 percent; Sao2 ranged from 94.5 to 95.7 percent; and Spo2 ranged from 93.7 to 96.3 percent (compared to 85–94 percent in healthy adults). Conclusions: In neonates, both left-shifted Hbo2 dissociation curve and lower AV differences of oxygen saturation measurements indicated low flow of oxygen to the body tissues. These findings demonstrate the importance of accurate assessment of oxygenation statues in neonates.

In these neonates, the mean AV blood differences for both So2 and Hbo2 were about 6 percent, which was much lower than those reported for healthy adults (23 percent) for O2 supply and demand. In addition, with very high levels of HbF releasing less O2 to the body tissue, the results of blood analyses are worrisome for these critically ill neonates for low systemic oxygen states.  O’Connor and Hall determined AV So2 in neonates without HbF determination. Much of the AV So2 difference is dependent on Svo2 measurement. The ranges of Svo2 spanned for 35 percent, and the ranges of Sao2 spanned 6 percent in these neonates. The greater intervals for Svo2 measurements contribute to greater sensitivity for the measurements (than Sao2 measurements) in responding to nursing care and changes of O2 demand. Thus, Svo2 measurement is essential for better assessment of oxygenation status in neonates.

The findings of this study on AV differences of So2 were limited with very small number of paired AV blood samples. However, critically ill neonates need accurate assessment of oxygenation status because of HbF, which releases less O2 to the tissues. Decreased differences of AV So2 measurements added further possibilities of lower flow of O2 to the body tissues and demonstrated the greater need to accurately assess the proper oxygenation in the neonates. The findings of this study continued to clarify the accuracy of So2 measurements for neonates. Additional studies are needed to examine So2 levels in neonates to further validate these findings by using larger sample sizes.

Neonatal ventilation strategies and long-term respiratory outcomes

Sandeep Shetty, Anne Greenough
Early Human Development 90 (2014) 735–739
http://dx.doi.org/10.1016/j.earlhumdev.2014.08.020

Long-term respiratory morbidity is common, particularly in those born very prematurely and who have developed bronchopulmonary dysplasia (BPD), but it does occur in those without BPD and in infants born at term. A variety of neonatal strategies have been developed, all with short-term advantages, but meta-analyses of randomized controlled trials (RCTs) have demonstrated that only volume-targeted ventilation and prophylactic high-frequency oscillatory ventilation (HFOV) may reduce BPD. Few RCTs have incorporated long-term follow-up, but one has demonstrated that prophylactic HFOV improves respiratory and functional outcomes at school age, despite not reducing BPD. Results from other neonatal interventions have demonstrated that any impact on BPD may not translate into changes in long-term outcomes. All future neonatal  ventilation RCTs should have long-term outcomes rather than BPD as their primary outcome if they are to impact on clinical practice.

A Model Analysis of Arterial Oxygen Desaturation during Apnea in Preterm Infants

Scott A. Sands, BA Edwards, VJ Kelly, MR Davidson, MH Wilkinson, PJ Berger
PLoS Comput Biol 5(12): e1000588
http://dx.doi.org:/10.1371/journal.pcbi.1000588

Rapid arterial O2 desaturation during apnea in the preterm infant has obvious clinical implications but to date no adequate explanation for why it exists. Understanding the factors influencing the rate of arterial O2 desaturation during apnea (_SSaO2 ) is complicated by the non-linear O2 dissociation curve, falling pulmonary O2 uptake, and by the fact that O2 desaturation is biphasic, exhibiting a rapid phase (stage 1) followed by a slower phase when severe desaturation develops (stage 2). Using a mathematical model incorporating pulmonary uptake dynamics, we found that elevated metabolic O2 consumption accelerates _SSaO2 throughout the entire desaturation process. By contrast, the remaining factors have a restricted temporal influence: low pre-apneic alveolar PO2 causes an early onset of desaturation, but thereafter has little impact; reduced lung volume, hemoglobin content or cardiac output, accelerates _SSaO2 during stage 1, and finally, total blood O2 capacity (blood volume and hemoglobin content) alone determines _SSaO2 during stage 2. Preterm infants with elevated metabolic rate, respiratory depression, low lung volume, impaired cardiac reserve, anemia, or hypovolemia, are at risk for rapid and profound apneic hypoxemia. Our insights provide a basic physiological framework that may guide clinical interpretation and design of interventions for preventing sudden apneic hypoxemia.

A novel approach to study oxidative stress in neonatal respiratory distress syndrome

Reena Negi, D Pande, K Karki, A Kumar, RS Khanna, HD Khanna
BBA Clinical 3 (2015) 65–69
http://dx.doi.org/10.1016/j.bbacli.2014.12.001

Oxidative stress is an imbalance between the systemic manifestation of reactive oxygen species and a biological system’s ability to readily detoxify the reactive intermediates or to repair the resulting damage. It is a physiological event in the fetal-to-neonatal transition, which is actually a great stress to the fetus. These physiological changes and processes greatly increase the production of free radicals, which must be controlled by the antioxidant defense system, the maturation of which follows the course of the gestation. This could lead to several functional alterations with important repercussions for the infants. Adequately mature and healthy infants are able to tolerate this drastic change in the oxygen concentration. A problem occurs when the intrauterine development is incomplete or abnormal. Preterm or intrauterine growth retarded (IUGR) and low birth weight neonates are typically of this kind. An oxidant/antioxidant imbalance in infants is implicated in the pathogenesis of the major complications of prematurity including respiratory distress syndrome (RDS), necrotizing enterocolitis (NEC), chronic lung disease, retinopathy of prematurity and intraventricular hemorrhage (IVH).

Background: Respiratory distress syndrome of the neonate (neonatal RDS) is still an important problem in treatment of preterm infants. It is accompanied by inflammatory processes with free radical generation and oxidative stress. The aim of study was to determine the role of oxidative stress in the development of neonatal RDS. Methods: Markers of oxidative stress and antioxidant activity in umbilical cord blood were studied in infants with neonatal respiratory distress syndrome with reference to healthy newborns. Results: Status of markers of oxidative stress (malondialdehyde, protein carbonyl and 8-hydroxy-2-deoxy guanosine) showed a significant increase with depleted levels of total antioxidant capacity in neonatal RDS when compared to healthy newborns. Conclusion: The study provides convincing evidence of oxidative damage and diminished antioxidant defenses in newborns with RDS. Neonatal RDS is characterized by damage of lipid, protein and DNA, which indicates the augmentation of oxidative stress. General significance: The identification of the potential biomarker of oxidative stress consists of a promising strategy to study the pathophysiology of neonatal RDS.

Neonatal respiratory distress syndrome represents the major lung complications of newborn babies. Preterm neonates suffer from respiratory distress syndrome (RDS) due to immature lungs and require assisted ventilation with high concentrations of oxygen. The pathogenesis of this disorder is based on the rapid formation of the oxygen reactive species, which surpasses the detoxification capacity of antioxidative defense system. The high chemical reactivity of free radical leads to damage to a variety of cellular macro molecules including proteins, lipids and nucleic acid. This results in cell injury and may induce respiratory cell death.

Malondialdehyde (MDA) is one of the final products of polyunsaturated fatty acids peroxidation. The present study showed increased concentration of MDA in neonates with respiratory disorders than that of control in consonance with the reported study.

Anemia, Apnea of Prematurity, and Blood Transfusions

Kelley Zagol, Douglas E. Lake, Brooke Vergales, Marion E. Moorman, et al
J Pediatr 2012;161:417-21
http://dx.doi.org:/10.1016/j.jpeds.2012.02.044

The etiology of apnea of prematurity is multifactorial; however, decreased oxygen carrying capacity may play a role. The respiratory neuronal network in neonates is immature, particularly in those born preterm, as demonstrated by their paradoxical response to hypoxemia. Although adults increase the minute ventilation in response to hypoxemia, newborns have a brief increase in ventilation followed by periodic breathing, respiratory depression, and occasionally cessation of respiratory effort. This phenomenon may be exacerbated by anemia in preterm newborns, where a decreased oxygen carrying capacity may result in decreased oxygen delivery to the central nervous system, a decreased efferent output of the respiratory neuronal network, and an increase in apnea.

Objective Compare the frequency and severity of apneic events in very low birth weight (VLBW) infants before and after blood transfusions using continuous electronic waveform analysis. Study design We continuously collected waveform, heart rate, and oxygen saturation data from patients in all 45 neonatal intensive care unit beds at the University of Virginia for 120 weeks. Central apneas were detected using continuous computer processing of chest impedance, electrocardiographic, and oximetry signals. Apnea was defined as respiratory pauses of >10, >20, and >30 seconds when accompanied by bradycardia (<100 beats per minute) and hypoxemia (<80% oxyhemoglobin saturation as detected by pulse oximetry). Times of packed red blood cell transfusions were determined from bedside charts. Two cohorts were analyzed. In the transfusion cohort, waveforms were analyzed for 3 days before and after the transfusion for all VLBW infants who received a blood transfusion while also breathing spontaneously. Mean apnea rates for the previous 12 hours were quantified and differences for 12 hours before and after transfusion were compared. In the hematocrit cohort, 1453 hematocrit values from all VLBW infants admitted and breathing spontaneously during the time period were retrieved, and the association of hematocrit and apnea in the next 12 hours was tested using logistic regression. Results Sixty-seven infants had 110 blood transfusions during times when complete monitoring data were available. Transfusion was associated with fewer computer-detected apneic events (P < .01). Probability of future apnea occurring within 12 hours increased with decreasing hematocrit values (P < .001). Conclusions Blood transfusions are associated with decreased apnea in VLBW infants, and apneas are less frequent at higher hematocrits.

Bronchopulmonary dysplasia: The earliest and perhaps the longest lasting obstructive lung disease in humans

Silvia Carraro, M Filippone, L Da Dalt, V Ferraro, M Maretti, S Bressan, et al.
Early Human Development 89 (2013) S3–S5
http://dx.doi.org/10.1016/j.earlhumdev.2013.07.015

Bronchopulmonary dysplasia (BPD) is one of the most important sequelae of premature birth and the most common form of chronic lung disease of infancy, an umbrella term for a number of different diseases that evolve as a consequence of a neonatal respiratory disorder. BPD is defined as the need for supplemental oxygen for at least 28 days after birth, and its severity is graded according to the respiratory support required at 36 post-menstrual weeks.

BPD was initially described as a chronic respiratory disease occurring in premature infants exposed to mechanical ventilation and oxygen supplementation. This respiratory disease (later named “old BPD”) occurred in relatively large premature newborn and, from a pathological standpoint, it was characterized by intense airway inflammation, disruption of normal pulmonary structures and lung fibrosis.

Bronchopulmonary dysplasia (BPD) is one of the most important sequelae of premature birth and the most common form of chronic lung disease of infancy. From a clinical standpoint BPD subjects are characterized by recurrent respiratory symptoms, which are very frequent during the first years of life and, although becoming less severe as children grow up, they remain more common than in term-born controls throughout childhood, adolescence and into adulthood. From a functional point of view BPD subjects show a significant airflow limitation that persists during adolescence and adulthood and they may experience an earlier and steeper decline in lung function during adulthood. Interestingly, patients born prematurely but not developing BPD usually fare better, but they too have airflow limitations during childhood and later on, suggesting that also prematurity per se has life-long detrimental effects on pulmonary function. For the time being, little is known about the presence and nature of pathological mechanisms underlying the clinical and functional picture presented by BPD survivors. Nonetheless, recent data suggest the presence of persistent neutrophilic airway inflammation and oxidative stress and it has been suggested that BPD may be sustained in the long term by inflammatory pathogenic mechanisms similar to those underlying COPD. This hypothesis is intriguing but more pathological data are needed.  A better understanding of these pathogenetic mechanisms, in fact, may be able to orient the development of novel targeted therapies or prevention strategies to improve the overall respiratory health of BPD patients.

We have a limited understanding of the presence and nature of pathological mechanisms in the lung of BPD survivors. The possible role of asthma-like inflammation has been investigated because BPD subjects often present with recurrent wheezing and other symptoms resembling asthma during their childhood and adolescence. But BPD subjects have normal or lower than normal exhaled nitric oxide levels and exhaled air temperatures, whereas they are higher than normal in asthmatic patients.

Of all obstructive lung diseases in humans, BPD has the earliest onset and is possibly the longest lasting. Given its frequent association with other conditions related to preterm birth (e.g. growth retardation, pulmonary hypertension, neurodevelopmental delay, hearing defects, and retinopathy of prematurity), it often warrants a multidisciplinary management.

Effects of Sustained Lung Inflation, a lung recruitment maneuver in primary acute respiratory distress syndrome, in respiratory and cerebral outcomes in preterm infants

Chiara Grasso, Pietro Sciacca, Valentina Giacchi, Caterina Carpinato, et al.
Early Human Development 91 (2015) 71–75
http://dx.doi.org/10.1016/j.earlhumdev.2014.12.002

Background: Sustained Lung Inflation (SLI) is a maneuver of lung recruitment in preterm newborns at birth that can facilitate the achieving of larger inflation volumes, leading to the clearance of lung fluid and formation of functional residual capacity (FRC). Aim: To investigate if Sustained Lung Inflation (SLI) reduces the need of invasive procedures and iatrogenic risks. Study design: 78 newborns (gestational age ≤ 34 weeks, weighing ≤ 2000 g) who didn’t breathe adequately at birth and needed to receive SLI in addition to other resuscitation maneuvers (2010 guidelines). Subjects: 78 preterm infants born one after the other in our department of Neonatology of Catania University from 2010 to 2012. Outcome measures: The need of intubation and surfactant, the ventilation required, radiological signs, the incidence of intraventricular hemorrhage (IVH), periventricular leukomalacia, retinopathy in prematurity from III to IV plus grades, bronchopulmonary dysplasia, patent ductus arteriosus, pneumothorax and necrotizing enterocolitis. Results: In the SLI group infants needed less intubation in the delivery room (6% vs 21%; p b 0.01), less invasive mechanical ventilation (14% vs 55%; p ≤ 0.001) and shorter duration of ventilation (9.1 days vs 13.8 days; p ≤ 0.001). There wasn’t any difference for nasal continuous positive airway pressure (82% vs 77%; p = 0.43); but there was less surfactant administration (54% vs 85%; p ≤ 0.001) and more infants received INSURE (40% vs 29%; p=0.17). We didn’t found any differences in the outcomes, except for more mild intraventricular hemorrhage in the SLI group (23% vs 14%; p = 0.15; OR= 1.83). Conclusion: SLI is easier to perform even with a single operator, it reduces the necessity of more complicated maneuvers and surfactant without statistically evident adverse effects.

Long-term respiratory consequences of premature birth at less than 32 weeks of gestation

Anne Greenough
Early Human Development 89 (2013) S25–S27
http://dx.doi.org/10.1016/j.earlhumdev.2013.07.004

Chronic respiratory morbidity is a common adverse outcome of very premature birth, particularly in infants who had developed bronchopulmonary dysplasia (BPD). Prematurely born infants who had BPD may require supplementary oxygen at home for many months and affected infants have increased healthcare utilization until school age. Chest radiograph abnormalities are common; computed tomography of the chest gives predictive information in children with ongoing respiratory problems. Readmission to hospital is common, particularly for those who have BPD and suffer respiratory syncytial virus lower respiratory infections (RSV LRTIs). Recurrent respiratory symptoms requiring treatment are common and are associated with evidence of airways obstruction and gas trapping. Pulmonary function improves with increasing age, but children with BPD may have ongoing airflow limitation. Lung function abnormalities may be more severe in those who had RSV LRTIs, although this may partly be explained by worse premorbid lung function. Worryingly, lung function may deteriorate during the first year. Longitudinal studies are required to determine if there is catch up growth.

Long-term pulmonary outcomes of patients with bronchopulmonary dysplasia

Anita Bhandari and Sharon McGrath-Morrow
Seminars in Perinatology 37 (2013)132–137
http://dx.doi.org/10.1053/j.semperi.2013.01.010

Bronchopulmonary dysplasia (BPD) is the commonest cause of chronic lung disease in infancy. The incidence of BPD has remained unchanged despite many advances in neonatal care. BPD starts in the neonatal period but its effects can persist long term. Premature infants with BPD have a greater incidence of hospitalization, and continue to have a greater respiratory morbidity and need for respiratory medications, compared to those without BPD. Lung function abnormalities, especially small airway abnormalities, often persist. Even in the absence of clinical symptoms, BPD survivors have persistent radiological abnormalities and presence of emphysema has been reported on chest computed tomography scans. Concern regarding their exercise tolerance remains. Long-term effects of BPD are still unknown, but given reports of a more rapid decline in lung function and their susceptibility to develop chronic obstructive pulmonary disease phenotype with aging, it is imperative that lung function of survivors of BPD be closely monitored.

Neonatal ventilation strategies and long-term respiratory outcomes

Sandeep Shetty, Anne Greenough
Early Human Development 90 (2014) 735–739
http://dx.doi.org/10.1016/j.earlhumdev.2014.08.020

Long-term respiratory morbidity is common, particularly in those born very prematurely and who have developed bronchopulmonary dysplasia (BPD), but it does occur in those without BPD and in infants born at term. A variety of neonatal strategies have been developed, all with short-term advantages, but meta-analyses of randomized controlled trials (RCTs) have demonstrated that only volume-targeted ventilation and prophylactic high-frequency oscillatory ventilation (HFOV) may reduce BPD. Few RCTs have incorporated long-term follow-up, but one has demonstrated that prophylactic HFOV improves respiratory and functional outcomes at school age, despite not reducing BPD. Results from other neonatal interventions have demonstrated that any impact on BPD may not translate into changes in long-term outcomes. All future neonatal ventilation RCTs should have long-term outcomes rather than BPD as their primary outcome if they are to impact on clinical practice.

Prediction of neonatal respiratory distress syndrome in term pregnancies by assessment of fetal lung volume and pulmonary artery resistance index

Mohamed Laban, GM Mansour, MSE Elsafty, AS Hassanin, SS EzzElarab
International Journal of Gynecology and Obstetrics 128 (2015) 246–250
http://dx.doi.org/10.1016/j.ijgo.2014.09.018

Objective: To develop reference cutoff values for mean fetal lung volume (FLV) and pulmonary artery resistance index (PA-RI) for prediction of neonatal respiratory distress syndrome (RDS) in low-risk term pregnancies. Methods: As part of a cross-sectional study, women aged 20–35 years were enrolled and admitted to a tertiary hospital in Cairo, Egypt, for elective repeat cesarean at 37–40 weeks of pregnancy between January 1, 2012, and July 31, 2013. FLV was calculated by virtual organ computer-aided analysis, and PA-RI was measured by Doppler ultrasonography before delivery. Results: A total of 80 women were enrolled. Neonatal RDS developed in 11 (13.8%) of the 80 newborns. Compared with neonates with RDS, healthy neonates had significantly higher FLVs (P b 0.001) and lower PA-RIs (P b 0.001). Neonatal RDS is less likely with FLV of at least 32 cm3 or PA-RI less than or equal to 0.74. Combining these two measures improved the accuracy of prediction. Conclusion: The use of either FLV or PA-RI predicted neonatal RDS. The predictive value increased when these two measures were combined

Pulmonary surfactant - a front line of lung host defense, 2003 JCI0318650.f2

Pulmonary surfactant – a front line of lung host defense, 2003 JCI0318650.f2

Pulmonary hypertension in bronchopulmonary dysplasia

Sara K.Berkelhamer, Karen K.Mestan, and Robin H. Steinhorn
Seminars In  Perinatology 37 (2013)124–131
http://dx.doi.org/10.1053/j.semperi.2013.01.009

Pulmonary hypertension (PH) is a common complication of neonatal respiratory diseases, including bronchopulmonary dysplasia (BPD), and recent studies have increased aware- ness that PH worsens the clinical course, morbidity and mortality of BPD. Recent evidence indicates that up to 18% of all extremely low-birth-weight infants will develop some degree of PH during their hospitalization, and the incidence rises to 25–40% of the infants with established BPD. Risk factors are not yet well understood, but new evidence shows that fetal growth restriction is a significant predictor of PH. Echocardiography remains the primary method for evaluation of BPD-associated PH, and the development of standardized screening timelines and techniques for identification of infants with BPD-associated PH remains an important ongoing topic of investigation. The use of pulmonary vasodilator medications, such as nitric oxide, sildenafil, and others, in the BPD population is steadily growing, but additional studies are needed regarding their long-term safety and efficacy.
An update on pharmacologic approaches to bronchopulmonary dysplasia

Sailaja Ghanta, Kristen Tropea Leeman, and Helen Christou
Seminars In Perinatology 37 (2013)115–123
http://dx.doi.org/10.1053/j.semperi.2013.01.008

Bronchopulmonary dysplasia (BPD) is the most prevalent long-term morbidity in surviving extremely preterm infants and is linked to increased risk of reactive airways disease, pulmonary hypertension, post-neonatal mortality, and adverse neurodevelopmental outcomes. BPD affects approximately 20% of premature newborns, and up to 60% of premature infants born before completing 26 weeks of gestation. It is characterized by the need for assisted ventilation and/or supplemental oxygen at 36 weeks postmenstrual age. Approaches to prevention and treatment of BPD have evolved with improved understanding of its pathogenesis. This review will focus on recent advancements and detail current research in pharmacotherapy for BPD. The evidence for both current and potential future experimental therapies will be reviewed in detail. As our understanding of the complex and multifactorial pathophysiology of BPD changes, research into these current and future approaches must continue to evolve.

Methylxanthines
Diuretics and bronchodilators
Corticosteroids
Macrolide antibiotics
Recombinant human Clara cell 10-kilodalton protein(rhCC10)
Vitamin A
Surfactant
Leukotriene receptor antagonist
Pulmonary vasodilators

Skeletal and Muscle

Skeletal Stem Cells in Space and Time

Moustapha Kassem and Paolo Bianco
Cell  Jan 15, 2015; 160: 17-19
http://dx.doi.org/10.1016/j.cell.2014.12.034

The nature, biological characteristics, and contribution to organ physiology of skeletal stem cells are not completely determined. Chan et al. and Worthley et al. demonstrate that a stem cell for skeletal tissues, and a system of more restricted, downstream progenitors, can be identified in mice and demonstrate its role in skeletal tissue maintenance and regeneration.

The groundbreaking concept that bone, cartilage, marrow adipocytes, and hematopoiesis-supporting stroma could originate from a common progenitor and putative stem cell was surprising at the time when it was formulated (Owen and Friedenstein, 1988). The putative stem cell, nonhematopoietic in nature, would be found in the postnatal bone marrow stroma, generate tissues previously thought of as foreign to each other, and support the turnover of tissues and organs that self-renew at a much slower rate compared to other tissues associated with stem cells (blood, epithelia). This concept also connected bone and bone marrow as parts of a single-organ system, implying their functional interplay. For many years, the evidence underpinning the concept has been incomplete.

While multipotency of stromal progenitors has been demonstrated by in vivo transplantation experiments, self-renewal, the defining property of a stem cell, has not been easily demonstrated until recently in humans (Sacchetti et al., 2007) and mice (Mendez-Ferrer et al., 2010). Meanwhile, a confusing and plethoric terminology has been introduced into the literature, which diverted and confounded the search for a skeletal stem cell and its physiological significance (Bianco et al., 2013).

Two studies in this issue of Cell (Chan et al., 2015; Worthley et al., 2015), using a combination of rigorous single-cell analyses and lineage tracing technologies, mark significant steps toward rectifying the course of skeletal stem cell discovery by making several important points, within and beyond skeletal physiology.

First, a stem cell for skeletal tissues, and a system of more restricted, downstream progenitors can in fact be identified and linked to defined phenotype(s) in the mouse. The system is framed conceptually, and approached experimentally, similar to the hematopoietic system.

Second, based on its assayable functions and potential, the stem cell at the top of the hierarchy is defined as a skeletal stem cell (SSC). As noted earlier (Sacchetti et al., 2007) (Bianco et al., 2013), this term clarifies, well beyond semantics, that the range of tissues that the self-renewing stromal progenitor (originally referred to as an ‘‘osteogenic’’ or ‘‘stromal’’ stem cell) (Owen and Friedenstein, 1988) can actually generate in vivo, overlaps with the range of tissues that make up the skeleton.

Third, these cells are spatially restricted, local residents of the bone/bone marrow organ. The systemic circulation is not a sizable contributor to their recruitment to locally deployed functions.

Fourth, a native skeletogenic potential is inherent to the system of progenitor/ stem cells found in the skeleton, and internally regulated by bone morphogenetic protein (BMP) signaling. This is reflected in the expression of regulators and antagonists of BMP signaling within the system, highlighting potential feedback mechanisms modulating expansion or quiescence of specific cell compartments.

Fifth, in cells isolated from other tissues, an assayable skeletogenic potential is not inherent: it can only be induced de novo by BMP reprogramming. These two studies (Chan et al., 2015, Worthley et al., 2015) corroborate the classical concept of ‘‘determined’’ and ‘‘inducible’’ skeletal progenitors (Owen and Friedenstein, 1988): the former residing in the skeleton, the latter found in nonskeletal tissues; the former capable of generating skeletal tissues, in vivo and spontaneously, the latter requiring reprogramming signals in order to acquire a skeletogenic capacity; the former operating in physiological bone formation, the latter in unwanted, ectopic bone formation in diseases such as fibrodysplasia ossificans progressiva.

To optimize our ability to obtain specific skeletal tissues for medical application, the study by Chan et al. offers a glimpse of another facet of the biology of SSC lineages and progenitors. Chan et al. show that a homogeneous cell population inherently committed to chondrogenesis can alter its output to generate bone if cotransplanted with multipotent progenitors. Conversely, osteogenic cells can be shifted to a chondrogenic fate by blockade of vascular endothelial growth factor receptor, consistent with the avascular and hypoxic milieu of cartilage. This has two important implications:

  • commitment is flexible in the system;
  • the choir is as important as the soloist and can modulate the solo tune.

Reversibility and population behavior thus emerge as two features that may be characteristic, albeit not unique, of the stromal system, resonating with conceptually comparable evidence in the human system.

The two studies by Chan et al. and Worthely et al. emphasize the relevance not only of their new data, but also of a proper concept of a skeletal stem cell per se, for proper clinical use. Confusion arising from improper conceptualization of skeletal stem cells has markedly limited clinical development of skeletal stem cell biology.

Gremlin 1 Identifies a Skeletal Stem Cell with Bone, Cartilage, and Reticular Stromal Potential

Daniel L. Worthley, Michael Churchill, Jocelyn T. Compton, Yagnesh Tailor, et al.
Cell, Jan 15, 2015; 160: 269–284
http://dx.doi.org/10.1016/j.cell.2014.11.042

The stem cells that maintain and repair the postnatal skeleton remain undefined. One model suggests that perisinusoidal mesenchymal stem cells (MSCs) give rise to osteoblasts, chondrocytes, marrow stromal cells, and adipocytes, although the existence of these cells has not been proven through fate-mapping experiments. We demonstrate here that expression of the bone morphogenetic protein (BMP) antagonist gremlin 1 defines a population of osteochondroreticular (OCR) stem cells in the bone marrow. OCR stem cells self-renew and generate osteoblasts, chondrocytes, and reticular marrow stromal cells, but not adipocytes. OCR stem cells are concentrated within the metaphysis of long bones not in the perisinusoidal space and are needed for bone development, bone remodeling, and fracture repair. Grem1 expression also identifies intestinal reticular stem cells (iRSCs) that are cells of origin for the periepithelial intestinal mesenchymal sheath. Grem1 expression identifies distinct connective tissue stem cells in both the bone (OCR stem cells) and the intestine (iRSCs).

Identification and Specification of the Mouse Skeletal Stem Cell

Charles K.F. Chan, Eun Young Seo, James Y. Chen, David Lo, A McArdle, et al.
Cell, Jan 15, 2015; 160: 285–298
http://dx.doi.org/10.1016/j.cell.2014.12.002

How are skeletal tissues derived from skeletal stem cells? Here, we map bone, cartilage, and stromal development from a population of highly pure, postnatal skeletal stem cells (mouse skeletal stem cells, mSSCs) to their downstream progenitors of bone, cartilage, and stromal tissue. We then investigated the transcriptome of the stem/progenitor cells for unique gene-expression patterns that would indicate potential regulators of mSSC lineage commitment. We demonstrate that mSSC niche factors can be potent inducers of osteogenesis, and several specific combinations of recombinant mSSC niche factors can activate mSSC genetic programs in situ, even in nonskeletal tissues, resulting in de novo formation of cartilage or bone and bone marrow stroma. Inducing mSSC formation with soluble factors and subsequently regulating the mSSC niche to specify its differentiation toward bone, cartilage, or stromal cells could represent a paradigm shift in the therapeutic regeneration of skeletal tissues.

Bone mesenchymal development

Bone mesenchymal development

Bone mesenchymal development

The bone-remodeling cycle

The bone-remodeling cycle

Nuclear receptor modulation – Role of coregulators in selective estrogen receptor modulator (SERM) actions

Qin Feng, Bert W. O’Malley
Steroids 90 (2014) 39–43
http://dx.doi.org/10.1016/j.steroids.2014.06.008

Selective estrogen receptor modulators (SERMs) are a class of small-molecule chemical compounds that bind to estrogen receptor (ER) ligand binding domain (LBD) with high affinity and selectively modulate ER transcriptional activity in a cell- and tissue-dependent manner. The prototype of SERMs is tamoxifen, which has agonist activity in bone, but has antagonist activity in breast. Tamoxifen can reduce the risk of breast cancer and, at same time, prevent osteoporosis in postmenopausal women. Tamoxifen is widely prescribed for treatment and prevention of breast cancer. Mechanistically the activity of SERMs is determined by the selective recruitment of coactivators and corepressors in different cell types and tissues. Therefore, understanding the coregulator function is the key to understanding the tissue selective activity of SERMs.

Hematopoietic

Hematopoietic Stem Cell Arrival Triggers Dynamic Remodeling of the Perivascular Niche

Owen J. Tamplin, Ellen M. Durand, Logan A. Carr, Sarah J. Childs, et al.
Cell, Jan 15, 2015; 160: 241–252
http://dx.doi.org/10.1016/j.cell.2014.12.032

Hematopoietic stem and progenitor cells (HSPCs) can reconstitute and sustain the entire blood system. We generated a highly specific transgenic reporter of HSPCs in zebrafish. This allowed us to perform high resolution live imaging on endogenous HSPCs not currently possible in mammalian bone marrow. Using this system, we have uncovered distinct interactions between single HSPCs and their niche. When an HSPC arrives in the perivascular niche, a group of endothelial cells remodel to form a surrounding pocket. This structure appears conserved in mouse fetal liver. Correlative light and electron microscopy revealed that endothelial cells surround a single HSPC attached to a single mesenchymal stromal cell. Live imaging showed that mesenchymal stromal cells anchor HSPCs and orient their divisions. A chemical genetic screen found that the compound lycorine promotes HSPC-niche interactions during development and ultimately expands the stem cell pool into adulthood. Our studies provide evidence for dynamic niche interactions upon stem cell colonization.

Neonatal anemia

Sanjay Aher, Kedar Malwatkar, Sandeep Kadam
Seminars in Fetal & Neonatal Medicine (2008) 13, 239e247
http://dx.doi.org:/10.1016/j.siny.2008.02.009

Neonatal anemia and the need for red blood cell (RBC) transfusions are very common in neonatal intensive care units. Neonatal anemia can be due to blood loss, decreased RBC production, or increased destruction of erythrocytes. Physiologic anemia of the newborn and anemia of prematurity are the two most common causes of anemia in neonates. Phlebotomy losses result in much of the anemia seen in extremely low birthweight infants (ELBW). Accepting a lower threshold level for transfusion in ELBW infants can prevent these infants being exposed to multiple donors.

Management of anemia in the newborn

Naomi L.C. Luban
Early Human Development (2008) 84, 493–498
http://dx.doi.org:/10.1016/j.earlhumdev.2008.06.007

Red blood cell (RBC) transfusions are administered to neonates and premature infants using poorly defined indications that may result in unintentional adverse consequences. Blood products are often manipulated to limit potential adverse events, and meet the unique needs of neonates with specific diagnoses. Selection of RBCs for small volume (5–20 mL/kg) transfusions and for massive transfusion, defined as extracorporeal bypass and exchange transfusions, are of particular concern to neonatologists. Mechanisms and therapeutic treatments to avoid transfusion are another area of significant investigation. RBCs collected in anticoagulant additive solutions and administered in small aliquots to neonates over the shelf life of the product can decrease donor exposure and has supplanted the use of fresh RBCs where each transfusion resulted in a donor exposure. The safety of this practice has been documented and procedures established to aid transfusion services in ensuring that these products are available. Less well established are the indications for transfusion in this population; hemoglobin or hematocrit alone are insufficient indications unless clinical criteria (e.g. oxygen desaturation, apnea and bradycardia, poor weight gain) also augment the justification to transfuse. Comorbidities increase oxygen consumption demands in these infants and include bronchopulmonary dysplasia, rapid growth and cardiac dysfunction. Noninvasive methods or assays have been developed to measure tissue oxygenation; however, a true measure of peripheral oxygen offloading is needed to improve transfusion practice and determine the value of recombinant products that stimulate erythropoiesis. The development of such noninvasive methods is especially important since randomized, controlled clinical trials to support specific practices are often lacking, due at least in part, to the difficulty of performing such studies in tiny infants.
The Effect of Blood Transfusion on the Hemoglobin Oxygen Dissociation Curve of Very Early Preterm Infants During the First Week of Life

Virginie De HaUeux, Anita Truttmann, Carmen Gagnon, and Harry Bard
Seminars in Perinatology, 2002; 26(6): 411-415
http://dx.doi.org:/10.1053/sper.2002.37313

This study was conducted during the first week of life to determine the changes in Ps0 (PO2 required to achieve a saturation of 50% at pH 7.4 and 37~ and the proportions of fetal hemoglobin (I-IbF) and adult hemoglobin (HbA) prior to and after transfusion in very early preterm infants. Eleven infants with a gestational age <–27 weeks have been included in study. The hemoglobin dissociation curve and the Ps0 was determined by Hemox-analyser. Liquid chromatography was also performed to determine the proportions of HbF and HbA. The mean gestational age of the 11 infants was 25.1 weeks (-+1 weeks) and their mean birth weight was 736 g (-+125 g). They received 26.9 mL/kg of packed red cells. The mean Ps0 prior and after transfusion was 18.5 +- 0.8 and 21.0 + 1 mm Hg (P = .0003) while the mean percentage of HbF was 92.9 -+ 1.1 and 42.6 -+ 5.7%, respectively. The data of this study show a decrease of hemoglobin oxygen affinity as a result of blood transfusion in very early preterm infants prone to O 2 toxicity. The shift in HbO 2 curve after transfusion should be taken into consideration when oxygen therapy is being regulated for these infants.

Effect of neonatal hemoglobin concentration on long-term outcome of infants affected by fetomaternal hemorrhage

Mizuho Kadooka, H Katob, A Kato, S Ibara, H Minakami, Yuko Maruyama
Early Human Development 90 (2014) 431–434
http://dx.doi.org/10.1016/j.earlhumdev.2014.05.010

Background: Fetomaternal hemorrhage (FMH) can cause severe morbidity. However, perinatal risk factors for long-term poor outcome due to FMH have not been extensively studied.                                                                                 Aims: To determine which FMH infants are likely to have neurological sequelae.
Study design: A single-center retrospective observational study. Perinatal factors, including demographic characteristics, Kleihauer–Betke test, blood gas analysis, and neonatal blood hemoglobin concentration ([Hb]), were analyzed in association with long-term outcomes.
Subjects: All 18 neonates referred to a Neonatal Intensive Care Unit of Kagoshima City Hospital and diagnosed with FMH during a 15-year study period. All had a neonatal [Hb] b7.5 g/dL and 15 of 17 neonates tested had Kleihauer–Betke test result N4.0%.
Outcome measures: Poor long-term outcome was defined as any of the following determined at 12 month old or more: cerebral palsy, mental retardation, attention deficit/hyperactivity disorder, and epilepsy.
Results: Nine of the 18 neonates exhibited poor outcomes. Among demographic characteristics and blood variables compared between two groups with poor and favorable outcomes, significant differences were observed in [Hb] (3.6 ± 1.4 vs. 5.4 ± 1.1 g/dL, P = 0.01), pH (7.09 ± 0.11 vs. 7.25 ± 0.13, P = 0.02) and base deficits (17.5 ± 5.4 vs. 10.4 ± 6.0 mmol/L, P = 0.02) in neonatal blood, and a number of infants with [Hb] ≤ 4.5 g/dL (78%[7/9] vs. 22%[2/9], P= 0.03), respectively. The base deficit in neonatal arterial blood increased significantly with decreasing neonatal [Hb].
Conclusions: Severe anemia causing severe base deficit is associated with neurological sequelae in FMH infants

Clinical and hematological presentation among Indian patients with common hemoglobin variants

Khushnooma Italia, Dipti Upadhye, Pooja Dabke, Harshada Kangane, et al.
Clinica Chimica Acta 431 (2014) 46–51
http://dx.doi.org/10.1016/j.cca.2014.01.028

Background: Co-inheritance of structural hemoglobin variants like HbS, HbD Punjab and HbE can lead to a variable clinical presentation and only few cases have been described so far in the Indian population.
Methods: We present the varied clinical and hematological presentation of 22 cases (HbSD Punjab disease-15, HbSE disease-4, HbD Punjab E disease-3) referred to us for diagnosis.
Results: Two of the 15 HbSDPunjab disease patients had moderate crisis, one presented with mild hemolytic anemia; however, the other 12 patients had a severe clinical presentation with frequent blood transfusion requirements, vaso occlusive crisis, avascular necrosis of the femur and febrile illness. The 4 HbSE disease patients had a mild to moderate presentation. Two of the 3 HbD Punjab E patients were asymptomatic with one patient’s sibling having a mild presentation. The hemoglobin levels of the HbSD Punjab disease patients ranged from 2.3 to 8.5 g/dl and MCV from 76.3 to 111.6 fl. The hemoglobin levels of the HbD Punjab E and HbSE patients ranged from 10.8 to 11.9 and 9.8 to 10.0 g/dl whereas MCV ranged from 67.1 to 78.2 and 74.5 to 76.0 fl respectively.
Conclusions: HbSD Punjab disease patients should be identified during newborn screening programs and managed in a way similar to sickle cell disease. Couple at risk of having HbSD Punjab disease children may be given the option of prenatal diagnosis in subsequent pregnancies.

Sickle cell anemia is the most common hemoglobinopathy seen across the world. It is caused by a point mutation in the 6th codon of the beta (β) globin gene leading to the substitution of the amino acid glutamic acid to valine. The sickle gene is frequently seen in Africa, some Mediterranean countries, India, Middle East—Saudi Arabia and North America. In India the prevalence of hemoglobin S (HbS) carriers varies from 2 to 40% among different population groups and HbS is mainly seen among the scheduled tribe, scheduled caste and other backward class populations in the western, central and parts of eastern and southern India. Sickle cell anemia has a variable clinical presentation in India with the most severe clinical presentation seen in central India whereas patients in the western region show a mild to moderate clinical presentation.

Hemoglobin D Punjab (HbD Punjab) (also known as HbD Los-Angeles, HbD Portugal, HbD North Carolina, D Oak Ridge and D Chicago) is another hemoglobin variant due to a point mutation in codon 121 of the β globin gene resulting in the substitution of the amino acid glutamic acid to glycine. It is a widely distributed hemoglobin with a relatively low prevalence of 0.86% in the Indo-Pak subcontinent, 1–3% in north-western India, 1–3% in the Black population in the Caribbean and North America and has also been reported among the English. It accounts for 55.6% of all the Hb variants seen in the Xenjiang province of China.

Hemoglobin E (HbE) is the most common abnormal hemoglobin in Southeast Asia. In India, the frequency ranges from 4% to 51% in the north eastern region and 3% to 4% in West Bengal in the east. The HbE mutation (β26 GAG→AAG) creates an alternative splice site and the βE chain is insufficiently synthesized, hence the phenotype of this disorder is that of a mild form of β thalassemia.

Though these 3 structural variants are prevalent in different regions of India, their interaction is increasingly seen in all states of the country due to migration of people to different regions for a better livelihood. There are very few reports on interaction of these commonly seen Hb variants and the phenotypic–genotypic presentation of these cases is important for genetic counseling and management.

HbF of patients with HbSD Punjab disease with variable clinical severity. The HbF values of 4 patients are not included as they were post blood transfusion

The genotypes of the patients were confirmed by restriction enzyme digestion and ARMS (Fig). Patients 1 to 15 were characterized as compound heterozygous for HbS and HbD Punjab whereas patients 16 to 19 were characterized as compound heterozygous for HbS and HbE. Patient nos. 20 to 22 were characterized as compound heterozygous for HbE and HbD Punjab.

Molecular characterization of HbS and HbDPunjab by restriction enzyme digestion and of HbE by ARMS.

Molecular characterization of HbS and HbDPunjab by restriction enzyme digestion and of HbE by ARMS.

Molecular characterization of HbS and HbDPunjab by restriction enzyme digestion and of HbE by ARMS.

The 3 common β globin gene variants of hemoglobin, HbS, HbE and HbD Punjab are commonly seen in India, with HbS having a high prevalence in the central belt and some parts of western, eastern and southern India, HbE in the eastern and north eastern region whereas HbD is mostly seen in the north western part of India. These hemoglobin variants have been reported in different population groups. However, with migration and intermixing of the different populations from different geographic regions, occasional cases of HbSD Punjab and HbSE are being reported. There are several HbD variants like HbD Punjab, HbD Iran, HbD Ibadan. However, of these only HbD Punjab interacts with HbS to form a clinically significant condition as the glutamine residue facilitates polymerization of HbS. HbD Iran and HbD Ibadan are non-interacting and produce benign conditions like the sickle cell trait. The first case of HbSD Punjab disease was a brother and sister considered to have atypical sickle cell disease in 1934. This family was further reinvestigated and reported as the first case of HbD Los Angeles which has the same mutation as the HbD Punjab. Serjeant et al. reported HbD Punjab in an English parent in 6 out of 11 HbSD-Punjab disease cases. This has been suggested to be due to the stationing of nearly 50,000 British troops on the Indian continent for a period of 200 y and the introduction into Britain of their Anglo-Indian children.

HbSD Punjab disease shows a similar pattern to HbS homozygous on alkaline hemoglobin electrophoresis but can be differentiated on acid agar gel electrophoresis and on HPLC. In HbSD Punjab disease cases, the peripheral blood films show anisocytosis, poikilocytosis, target cells and irreversibly sickled cells. Values of HbF and HbA2 are similar to those in sickle homozygous cases. HbSD Punjab disease is characterized by a moderately severe hemolytic anemia.

Twenty-one cases of HbSDPunjab were reported by Serjeant of which 16 were reported by different workers among patients originating from Caucasian, Spanish, Australian, Irish, English, Portuguese, Black, American, Venezuelan, Caribbean, Mexican, Turkish and Jamaican backgrounds. Yavarian et al. 2009 reported a multi centric origin of HbD Punjab which in combination with HbS results in sickle cell disease. Patel et al. 2010 have also reported 12 cases of HbSD Punjab from the Orissa state of eastern India. Majority of these cases were symptomatic, presenting with chronic hemolytic anemia and frequent painful crises.

HbF levels >20% were seen in 4 out of our 11 clinically severe patients of HbSD-Punjab disease with the mean HbF levels of 16.8% in 8 clinically severe patients, while 3 clinically severe patients were post transfused. However, the 3 patients with a mild to moderate clinical presentation showed a mean HbF level of 8.6%. This is in contrast to the relatively milder clinical presentation associated with high HbF seen in patients with sickle cell anemia. This was also reported by Adekile et al. 2010 in 5 cases of HbS-DLos Angeles where high HbF did not ameliorate the severe clinical presentation seen in these patients.

These 15 cases of HbSDPunjab disease give us an overall idea of the severe clinical presentation of the disease in different regions of India. However the HbDPunjabE cases were milder or asymptomatic and the HbSE cases were moderately symptomatic. Since most of the cases of HbSDPunjab disease were clinically severe, it is important to pick up these cases during newborn screening and enroll them into a comprehensive care program with the other sickle cell disease patients with introduction of therapeutic interventions such as penicillin prophylaxis if required and pneumococcal immunization. In fact, 2 of our cases (No. 6 and 7) were identified during newborn screening for sickle cell disorders. The parents can be given information on home care and educated to detect symptoms that may lead to serious medical emergencies. The parents of these patients as well as the couples who are at risk of having a child with HbSDPunjab disease could also be counseled about the option of prenatal diagnosis in subsequent pregnancies. It is thus important to document the clinical and hematological presentation of compound heterozygotes with these common β globin chain variants.

Common Hematologic Problems in the Newborn Nursery

Jon F. Watchko
Pediatr Clin N Am – (2015) xxx-xxx
http://dx.doi.org/10.1016/j.pcl.2014.11.011

Common RBC disorders include hemolytic disease of the newborn, anemia, and polycythemia. Another clinically relevant hematologic issue in neonates to be covered herein is thrombocytopenia. Disorders of white blood cells will not be reviewed.

KEY POINTS

(1)               Early clinical jaundice or rapidly developing hyperbilirubinemia are often signs of hemolysis, the differential diagnosis of which commonly includes immune-mediated disorders, red-cell enzyme deficiencies, and red-cell membrane defects.

(2)             Knowledge of the maternal blood type and antibody screen is critical in identifying non-ABO alloantibodies in the maternal serum that may pose a risk for severe hemolytic disease in the newborn.

(3)             Moderate to severe thrombocytopenia in an otherwise well-appearing newborn strongly suggests immune-mediated (alloimmune or autoimmune) thrombocytopenia.

Hemolytic conditions in the neonate

1. Immune-mediated (positive direct Coombs test)  a. Rhesus blood group: Anti-D, -c, -C, -e, -E, CW, and several others

  b. Non-Rhesus blood groups: Kell, Duffy, Kidd, Xg, Lewis, MNS, and others

  c. ABO blood group: Anti-A, -B

2. Red blood cell (RBC) enzyme defects

  a. Glucose-6-phosphate dehydrogenase (G6PD) deficiency

  b. Pyruvate kinase deficiency

  c. Others

3. RBC membrane defects

  a. Hereditary spherocytosis

  b. Elliptocytosis

  c. Stomatocytosis

  d. Pyknocytosis

  e. Others

4. Hemoglobinopathies

  a. alpha-thalassemia

  b. gamma-thalassemia

Standard maternal antibody screeningAlloantibody                                 Blood Group

D, C, c, E, e, f, CW, V                     Rhesus

K, k, Kpa, Jsa                                  Kell

Fya, Fyb                                          Duffy

Jka, Jkb                                           Kidd

Xga                                                  Xg

Lea, Leb                                          Lewis

S, s, M, N                                        MNS

P1                                                    P

Lub                                                  Lutheran

Non-ABO alloantibodies reported to cause moderate to severe hemolytic disease of the newbornWithin Rh system: Anti-D, -c, -C, -Cw, -Cx, -e, -E, -Ew, -ce, -Ces, -Rh29, -Rh32, -Rh42, -f, -G, -Goa, -Bea, -Evans, -Rh17, -Hro, -Hr, -Tar, -Sec, -JAL, -STEM

Outside Rh system:  Anti-LW, -K, -k, -Kpa, -Kpb, -Jka, -Jsa, -Jsb, -Ku, -K11, -K22, -Fya, -M, -N, -S, -s, -U, -PP1 pk, -Dib, -Far, -MUT, -En3, -Hut, -Hil, -Vel, -MAM, -JONES, -HJK, -REIT

 

Red Blood Cell Enzymopathies

G6PD9 and pyruvate kinase (PK) deficiency are the 2 most common red-cell enzyme disorders associated with marked neonatal hyperbilirubinemia. Of these, G6PD deficiency is the more frequently encountered and it remains an important cause of kernicterus worldwide, including the United States, Canada, and the United Kingdom, the prevalence in Western countries a reflection in part of immigration patterns and intermarriage. The risk of kernicterus in G6PD deficiency also relates to the potential for unexpected rapidly developing extreme hyperbilirubinemia in this disorder associated with acute severe hemolysis.

Red Blood Cell Membrane Defects

Establishing a diagnosis of RBC membrane defects is classically based on the development of Coombs-negative hyperbilirubinemia, a positive family history, and abnormal RBC smear, albeit it is often difficult because newborns normally exhibit a marked variation in red-cell membrane size and shape. Spherocytes, however, are not often seen on RBC smears of hematologically normal newborns and this morphologic abnormality, when prominent, may yield a diagnosis of hereditary spherocytosis (HS) in the immediate neonatal period. Given that approximately 75% of families affected with hereditary spherocytosis manifest an autosomal dominant phenotype, a positive family history can often be elicited and provide further support for this diagnosis. More recently, Christensen and Henry highlighted the use of an elevated mean corpuscular hemoglobin concentration (MCHC) (>36.0 g/dL) and/or elevated ratio of MCHC to mean corpuscular volume, the latter they term the “neonatal HS index” (>0.36, likely >0.40) as screening tools for HS. An index of greater than 0.36 had 97% sensitivity, greater than 99% specificity, and greater than 99% negative predictive value for identifying HS in neonates. Christensen and colleagues also provided a concise update of morphologic RBC features that may be helpful in diagnosing this and other underlying hemolytic conditions in newborns.

The diagnosis of HS can be confirmed using the incubated osmotic fragility test when coupled with fetal red-cell controls or eosin-5-maleimide flow cytometry. One must rule out symptomatic ABO hemolytic disease by performing a direct Coombs test, as infants so affected also may manifest prominent micro-spherocytosis. Moreover, HS and symptomatic ABO hemolytic disease can occur in the same infant and result in severe hyperbilirubinemia and anemia.  Of other red-cell membrane defects, only hereditary elliptocytosis,  stomato-cytosis, and infantile pyknocytosis have been reported to exhibit significant hemolysis in the newborn period. Hereditary elliptocytosis and stomatocytosis are both rare. Infantile pyknocytosis, a transient red-cell membrane abnormality manifesting itself during the first few months of life, is more common.

Risk factors for bilirubin neurotoxicityIsoimmune hemolytic disease

G6PD deficiency

Asphyxia

Sepsis

Acidosis

Albumin less than 3.0 g/dL
Data from Maisels MJ, Bhutani VK, Bogen D, et al. Hyperbilirubinemia in the newborn infant > or 535 weeks’ gestation: an update with clarifications. Pediatrics 2009; 124:1193–8.

Polycythemia

Polycythemia (venous hematocrit 65%) in seen in infants across a range of conditions associated with active erythropoiesis or passive transfusion.76,77 They include, among others, placental insufficiency, the infant of a diabetic mother, recipient in twin-twin transfusion syndrome, and several aneuploidies, including trisomy. The clinical concern related to polycythemia is the risk for microcirculatory complications of hyperviscosity. However, determining which polycythemic infants are hyperviscous and when to intervene is a challenge.

 

 

Liver

Metabolic disorders presenting as liver disease

Germaine Pierre, Efstathia Chronopoulou
Paediatrics and Child Health 2013; 23(12): 509-514
The liver is a highly metabolically active organ and many inherited metabolic disorders have hepatic manifestations. The clinical presentation in these patients cannot usually be distinguished from liver disease due to acquired causes like infection, drugs or hematological disorders. Manifestations include acute and chronic liver failure, cholestasis and hepatomegaly. Metabolic causes of acute liver failure in childhood can be as high as 35%. Certain disorders like citrin deficiency and Niemann-Pick C disease may present in infancy with self-limiting cholestasis before presenting in later childhood or adulthood with irreversible disease. This article reviews important details from the history and clinical examination when evaluating the pediatric patient with suspected metabolic disease, the specialist and genetic tests when investigating, and also discusses specific disorders, their clinical course and treatment. The role of liver transplantation is also briefly discussed. Increased awareness of this group of disorders is important as in many cases, early diagnosis leads to early intervention with improved outcome. Diagnosis also allows genetic counselling and future family planning.

Adult liver disorders caused by inborn errors of metabolism: Review and update

Sirisak Chanprasert, Fernando Scaglia
Molecular Genetics and Metabolism 114 (2015) 1–10
http://dx.doi.org/10.1016/j.ymgme.2014.10.011

Inborn errors of metabolism (IEMs) are a group of genetic diseases that have protean clinical manifestations and can involve several organ systems. The age of onset is highly variable but IEMs afflict mostly the pediatric population. However, in the past decades, the advancement in management and new therapeutic approaches have led to the improvement in IEM patient care. As a result, many patients with IEMs are surviving into adulthood and developing their own set of complications. In addition, some IEMs will present in adulthood. It is important for internists to have the knowledge and be familiar with these conditions because it is predicted that more and more adult patients with IEMs will need continuity of care in the near future. The review will focus on Wilson disease, alpha-1 antitrypsin deficiency, citrin deficiency, and HFE-associated hemochromatosis which are typically found in the adult population. Clinical manifestations and pathophysiology, particularly those that relate to hepatic disease as well as diagnosis and management will be discussed in detail.

Inborn errors of metabolism (IEMs) are a group of genetic diseases characterized by abnormal processing of biochemical reactions, resulting in accumulation of toxic substances that could interfere with normal organ functions, and failure to synthesize essential compounds. IEMs are individually rare, but collectively numerous. The clinical presentations cover a broad spectrum and can involve almost any organ system. The age of onset is highly variable but IEMs afflict mostly the pediatric population.

Wilson disease is an autosomal recessive genetic disorder of copper metabolism. It is characterized by an abnormal accumulation of inorganic copper in various tissues, most notably in the liver and the brain, especially in the basal ganglia. The disease was first described in 1912 by Kinnier Wilson, and affects between 1 in 30,000 and 1 in 100,000 individuals. Clinical features are variable and depend on the extent  and the severity of copper deposition. Typically, patients tend to develop hepatic disease at a younger age than the neuropsychiatric manifestations. Individuals withWilson disease eventually succumb to complications of end stage liver disease or become debilitated from neurological problems, if they are left untreated.

The clinical presentations of Wilson disease are varied affecting many organ systems. However, the overwhelming majority of cases display hepatic and neurologic symptoms. In general, patients with hepatic disease present between the first and second decades of life although patients as young as 3 years old or over 50 years old have also been reported. The most common modes of presentations are acute self-limited hepatitis and chronic active hepatitis that are indistinguishable from other hepatic disorders although liver aminotransferases are generally much lower than in autoimmune or viral hepatitis. Acute fulminant hepatic failure is less common but is observed in approximately 3% of all cases of acute liver failure. Symptoms of acute liver failure include jaundice, coagulopathy, and hepatic encephalopathy. Cirrhosis can develop over time and may be clinically silent. Hepatocellular carcinoma (HCC) is rarely associated with Wilson disease, but may occur in the setting of cirrhosis and chronic inflammation.

Copper is an essential element, and is required for the proper functioning of various proteins and enzymes. The total body content of copper in a healthy adult individual is approximately 70–100 mg, while the daily requirements are estimated to be between 1 and 5 mg. Absorption occurs in the small intestine. Copper is taken up to the hepatocytes via the copper transporter hTR1. Once inside the cell, copper is bound to various proteins including metallothionein and glutathione, however, it is the metal chaperone, ATOX1 that helps direct copper to the ATP7B protein for intracellular transport and excretion. At the steady state, copper will be bound to ATP7B and is then incorporated to ceruloplasmin and secreted into the systemic circulation. When the cellular copper concentration arises, ATP7B protein will be redistributed from the trans-Golgi network to the prelysosomal vesicles facilitating copper excretion into the bile. The molecular defects in ATP7B lead to a reduction of copper excretion. Excess copper is accumulated in the liver causing tissue injury. The rate of accumulation of copper varies among individuals, and it may depend on other factors such as alcohol consumption, or viral hepatitis infections. If the liver damage is not severe, patients will accumulate copper in various tissues including the brain, the kidney, the eyes, and the musculoskeletal system leading to clinical disease. A failure of copper to incorporate into ceruloplasmin leads to secretion of the unsteady protein that has a shorter half-life, resulting in the reduced concentrations of ceruloplasmin seen in most patients with Wilson disease.

Wilson disease used to be a progressive fatal condition during the first half of the 20th century because there was no effective treatment available at that time. Penicillamine was the first pharmacologic agent introduced in 1956 for treating this condition. Penicillamine is a sulfhydryl-bearing amino acid cysteine doubly substituted with methyl groups. This drug acts as a chelating agent that promotes the urinary excretion of copper. It is rapidly absorbed in the gastrointestinal track, and over 80% of circulating penicillamine is excreted via the kidneys. Although it is very effective, approximately 10%–50% of Wilson disease patients with neuropsychiatric presentations may experience worsening of their symptoms, and often times the worsening symptoms may not be reversible.

Alpha1-antitrypsin deficiency

Alpha1-antitrypsin deficiency (AATD) is one of the most common genetic liver diseases in children and adults, affecting 1 in 2000 to 1 in 3000 live births worldwide. It is transmitted in an autosomal co-dominant fashion with variable expressivity. Alpha1 antitrypsin (A1AT) is a member of the serine protease inhibitor (SERPIN) family. Its function is to counteract the proteolytic effect of neutrophil elastase and other neutrophil proteases. Mutations in the SERPINA1, the gene encoding A1AT, result in changes in the protein structure with the PiZZ phenotype being the most common cause of liver and lung disease-associated AATDs. Although, it classically causes early onset chronic obstructive pulmonary disease (COPD) in adults, liver disease characterized by chronic inflammation, hepatic fibrosis, and cirrhosis is not uncommon in the adult population. Decreased plasma concentration of A1AT predisposes lung tissue to be more susceptible to injury from protease enzymes. However, the underlying mechanism of liver injury is different, and is believed to be caused by accumulation of polymerized mutant A1AT in the hepatocyte endoplasmic reticulum (ER). Currently, there is no specific treatment for liver disease-associated AATD, but A1AT augmentation therapy is available for patients affected with pulmonary involvement.

A1AT is a single-chain, 52-kDa polypeptide of approximately 394 amino acids [56]. It is synthesized in the liver, circulates in the plasma, and functions as an inhibitor of neutrophil elastase and other proteases such as cathepsin G, and proteinase 3. A1AT has a globular shape composed of two central β sheets surrounded by a small β sheet and nine α helices. The pathophysiology underlying liver disease is thought to be a toxic gain-of-function mutation associated with the PiZZ phenotypes. This hypothesis has been supported by the fact that null alleles which produce no detectable plasma A1AT, are not associated with liver disease. In addition, the transgenic mouse model of AATD PiZZ developed periodic acid-Schiff-positive diastase-resistant intrahepatic globule early in life similar to AATD patients. The PiZZ phenotype results in the blockade of the final processing of A1AT in the liver, as only 15% of the A1AT reaches the circulation whereas 85% of non-secreted protein is accumulated in the hepatocytes.

Citrin deficiency

Citrin deficiency is a relatively newly-defined autosomal recessive disease. It encompasses two different sub-groups of patients, neonatal intrahepatic cholestasis caused by citrin deficiency (NICCD), and adult onset citrullinemia type 2 (CTLN 2).

AGC2 exports aspartate out of the mitochondrial matrix in exchange for glutamate and a proton. Thus, this protein has an important role in ureagenesis and gluconeogenesis. In CTLN2, a defect in this protein is believed to limit the supply of aspartate for the formation of argininosuccinate in the cytosol resulting in impairment of ureagenesis. Interestingly, the mouse model of citrin deficiency (Ctrn−/−) fails to develop symptoms of CTLN2 suggesting that the mitochondrial aspartate is not the only source of ureagenesis. However, it should be noted that the rodent liver expresses higher glycerol-phosphate shuttle activity than the human counterpart. With the intact glycerol-phosphate dehydrogenase, it can compensate for the deficiency of AGC2, as demonstrated by the AGC2 and glycerol-phosphate dehydrogenase double knock-out mice that exhibit similar features to those observed in human CTLN2.

HFE-associated hemochromatosis

HFE-associated hemochromatosis is an inborn error of iron metabolism characterized by excessive iron storage resulting in tissue and organ damage. It is the most common autosomal recessive disorder in the Caucasian population, affecting 0.3%–0.5% of individuals of Northern European descent. The term “hemochromatosis” was coined in 1889 by the German pathologist Friedrich Daniel Von Recklinghausen, who described it as bronze stain of organs caused by a blood borne pigment.

The classic clinical triad of cirrhosis, diabetes, and bronze skin pigmentation is rarely observed nowadays given the early recognition, diagnosis, and treatment of this condition. The most common presenting symptoms are nonspecific including weakness, lethargy, and arthralgia.

The liver is a major site of iron storage in healthy individuals and as such it is the organ that is universally affected in HFE-associated hemochromatosis. Elevation of liver aminotransferases indicative of hepatocyte injury is the most common mode of presentation and it can be indistinguishable from other causes of hepatitis. Approximately 15%–40% of patients with HFE-associated hemochromatosis have other liver conditions, including chronic viral hepatitis B or C infection, nonalcoholic fatty liver disease, and alcoholic liver disease.

 

The liver in haemochromatosis

Rune J. Ulvik
Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx
http://dx.doi.org/10.1016/j.jtemb.2014.08.005

The review deals with genetic, regulatory and clinical aspects of iron homeostasis and hereditary hemochromatosis. Hemochromatosis was first described in the second half of the 19th century as a clinical entity characterized by excessive iron overload in the liver. Later, increased absorption of iron from the diet was identified as the pathophysiological hallmark. In the 1970s genetic evidence emerged supporting the apparent inheritable feature of the disease. And finally in 1996 a new “hemochromato-sis gene” called HFE was described which was mutated in about 85% of the patients. From the year2000 onward remarkable progress was made in revealing the complex molecular regulation of iron trafficking in the human body and its disturbance in hemochromatosis. The discovery of hepcidin and ferroportin and their interaction in regulating the release of iron from enterocytes and macrophages to plasma were important milestones. The discovery of new, rare variants of non-HFE-hemochromatosis was explained by mutations in the multicomponent signal transduction pathway controlling hepcidin transcription. Inhibited transcription induced by the altered function of mutated gene products, results in low plasma levels of hepcidin which facilitate entry of iron from enterocytes into plasma. In time this leads to progressive accumulation of iron and subsequently development of disease in the liver and other parenchymatous organs. Being the major site of excess iron storage and hepcidin synthesis the liver is a cornerstone in maintaining normal systemic iron homeostasis. Its central pathophysiological role in HFE-hemochromatosis with downgraded hepcidin synthesis, was recently shown by the finding that liver transplantation normalized the hepcidin levels in plasma and there was no sign of iron accumulation in the new liver.

Gastrointestinal

Decoding the enigma of necrotizing enterocolitis in premature infants

Roberto Murgas TorrazzaNan Li, Josef Neu
Pathophysiology 21 (2014) 21–27
http://dx.doi.org/10.1016/j.pathophys.2013.11.011

Necrotizing enterocolitis (NEC) is an enigmatic disease that affects primarily premature infants. It often occurs suddenly and when it occurs, treatment attempts at treatment often fail and results in death. If the infant survives, there is a significant risk of long term sequelae including neurodevelopmental delays. The pathophysiology of NEC is poorly understood and thus prevention has been difficult. In this review, we will provide an overview of why progress may be slow in our understanding of this disease, provide a brief review diagnosis, treatment and some of the current concepts about the pathophysiology of this disease.

Necrotizing enterocolitis (NEC) has been reported since special care units began to house preterm infants .With the advent of modern neonatal intensive care approximately 40 years ago, the occurrence and recognition of the disease markedly increased. It is currently the most common and deadly gastro-intestinal illness seen in preterm infants. Despite major efforts to better understand, treat and prevent this devastating disease, little if any progress has been made during these 4 decades. Underlying this lack of progress is the fact that what is termed “NEC” is likely more than one disease, or mimicked by other diseases, each with a different etiopathogenesis.

Human gut microbiome

Human gut microbiome

Term or near term infants with “NEC” when compared to matched controls usually have occurrence of their disease in the first week after birth, have a significantly higher frequency of prolonged rupture of membranes, chorio-amnionitis, Apgar score <7 at 1 and 5 min, respiratory problems, congenital heart disease, hypoglycemia, and exchange transfusions. When a “NEC” like illness presents in term or near term infants, it should be noted that these are likely to be distinct in pathogenesis than the most common form of NEC and should be differentiated as such.

The infants who suffer primary ischemic necrosis are term or near term infants (although this can occur in preterms) who have concomitant congenital heart disease, often related to poor left ventricular output or obstruction. Other factors that have been associated with primary ischemia are maternal cocaine use, hyperviscosity caused by polycythemia or a severe antecedent hypoxic–ischemic event. Whether the dis-ease entity that results from this should be termed NEC can be debated on historical grounds, but the etiology is clearly different from the NEC seen in most preterm infants.

The pathogenesis of NEC is uncertain, and the etiology seems to be multifactorial. The “classic” form of NEC is highly associated with prematurity; intestinal barrier immaturity, immature immune response, and an immature regulation of intestinal blood flow (Fig.). Although genetics appears to play a role, the environment, especially a dysbiotic intestinal microbiota acting in concert with host immaturities predisposes the preterm infant to disruption of the intestinal epithelia, increased permeability of tight junctions, and release of inflammatory mediators that leads to intestinal mucosa injury and therefore development of necrotizing enterocolitis.

NEC is a multifactorial disease

NEC is a multifactorial disease

What causes NEC? NEC is a multifactorial disease with an interaction of several etiophathologies

It is clear from this review that there are several entities that have been described as NEC. What is also clear is that despite having some overlap in the final parts of the pathophysiologic cascade that lead to necrosis, the disease that is most commonly seen in the preterm infant is likely to have an origin that differs markedly from that seen in term infants with congenital heart disease or severe hypoxic–ischemic injury. Thus, epidemiologic studies will need to differentiate these entities, if the aim is to dissect common features that are most highly associated with development of the disease. At this juncture, we areleft with more of a population based preventative approach, where the use of human milk, evidence based feeding guide-lines, considerations for microbial therapy once these are proved safe and effective and approved as such by regulatory authorities, and perhaps even measures that prevent prematurity will have a major impact on this devastating disease.

Influenced by the microbiota, intestinal epithelial cells (IECs) elaborate cytokines

Influenced by the microbiota, intestinal epithelial cells (IECs) elaborate cytokines

Influenced by the microbiota, intestinal epithelial cells (IECs) elaborate cytokines, including thymic stromal lymphoprotein (TSLP), transforming growthfactor (TGF), and interleukin-10 (IL-10), that can influence pro-inflammatory cytokine production by dendritic cells (DC) and macrophages present in the laminapropria (GALT) and Peyer’s patches. Signals from commensal organisms may influence tissue-specific functions, resulting in T-cell expansion and regulation of the numbers of Th-1,
Th-2, and Th-3 cells. Also modulated by the microbiota, other IEC derived factors, including APRIL (a proliferation-inducing ligand),B-cell activating factor (BAFF), secretory leukocyte peptidase inhibitor (SLPI), prostaglandin E2(PGE2), and other metabolites, directly regulate functions ofboth antigen presenting cells and lymphocytes in the intestinal ecosystem. NK: natural killer cell; LN: lymph node; DC: dendritic cells.Modified from R. Sharma, C. Young, M. Mshvildadze, J. Neu, Intestinal microbiota does it play a role in diseases of the neonate? NeoReviews 10 (4) (2009)e166, with permission

Cross-talk between monocyte.macrophage cells and T.NK lymphocytes

Cross-talk between monocyte.macrophage cells and T.NK lymphocytes

Current Issues in the Management of Necrotizing Enterocolitis

Marion C. W. Henry and R. Lawrence Moss
Seminars in Perinatology, 2004; 28(3): 221-233
http://dx.doi.org:/10.1053/j.semperi.2004.03.010

Necrotizing enterocolitis is almost exclusively a disease of prematurity, with 90% of all cases occurring in premature infants and 90% of those infants weighing less than 2000 g. Prematurity is the only risk factor for necrotizing enterocolitis consistently identified in case control studies and the disease is rare in countries where prematurity is uncommon such as Japan and Sweden. When necrotizing enterocolitis does occur in full-term infants, it appears to by a somewhat different disease, typically associated with some predisposing condition.

NEC occurs in one to three in 1,000 live births and most commonly affects babies born between 30-32 weeks. It is most often diagnosed during the second week of life and occurs more often in previously fed infants. The mortality from NEC has been cited as 10% to 50% of all NEC cases. Surgical mortality has decreased over the last several decades from 70% to between 20 and 50%. The incremental cost per case of acute hospital care is estimated at $74 to 186 thousand compared to age matched controls, not including additional costs of long term care for the infants’ with lifelong morbidity. Survivors may develop short bowel syndrome, recurrent bouts of catheter-related sepsis, malabsorption, malnutrition, and TPN induced liver failure.

Although extensive research concerning the pathophysiology of necrotizing enterocolitis has occurred, a complete understanding has not been fully elucidated. The classic histologic finding is coagulation necrosis; present in over 90% of specimens. This finding suggests the importance of ischemia in the pathogenesis of NEC. Inflammation and bacterial overgrowth also are present. These findings support the assumptions by Kosloske that NEC occurs by the interaction of 3 events:

  • intestinal ischemia,
  • colonization by pathogenic bacteria and
  • excess protein substrate in the intestinal lumen.

Additionally, the immunologic immaturity of the neonatal gut has been implicated in the development of NEC. Reparative tissue changes including epithelial regeneration, formation of granulation tissue and fibrosis, and mixed areas of acute and chronic inflammatory changes suggest that the pathogenesis of NEC may involve a chronic process of injury and repair.

Premature newborns born prior to the 32nd week of gestational age may have compromised intestinal peristalsis and decreased motility. These motility problems may lead to poor clearance of bacteria, and subsequent bacterial overgrowth. Premature infants also have an immature intestinal tract in terms of immunologic immunity.

There are fewer functional B lymphocytes present and the ability to produce sufficient secretory IgA is reduced. Pepsin, gastric acid and mucus are also not produced as well in prematurity. All of these factors may contribute to the limited proliferation of intestinal flora and the decreased binding of these flora to mucosal cells (Fig).

Role of nitric oxide in the pathogenesis of NEC

Role of nitric oxide in the pathogenesis of NEC

Role of nitric oxide in the pathogenesis of NEC.

Characteristics of the immature gut leading to increased risk of necrotizing enterocolitis

Characteristics of the immature gut leading to increased risk of necrotizing enterocolitis

Characteristics of the immature gut leading to increased risk of necrotizing enterocolitis.

As understanding of the pathophysiology of necrotizing enterocolitis continues to evolve, a unifying concept is emerging. Initially, there is likely a subclinical insult leading to NEC. This may arise from a brief episode of hypoxia or infection. With colonization of the intestines, bacteria bind to the injured mucosa eliciting an inflammatory response which leads to further inflammation.

Intestinal Microbiota Development in Preterm Neonates and Effect of Perinatal Antibiotics

Silvia Arboleya, Borja Sanchez,, Christian Milani, Sabrina Duranti, et al.
Pediatr 2014;-:—).  http://dx.doi.org/10.1016/j.jpeds.2014.09.041

Objectives Assess the establishment of the intestinal microbiota in very low birth-weight preterm infants and to evaluate the impact of perinatal factors, such as delivery mode and perinatal antibiotics.
Study design We used 16S ribosomal RNA gene sequence-based microbiota analysis and quantitative polymerase chain reaction to evaluate the establishment of the intestinal microbiota. We also evaluated factors affecting the microbiota, during the first 3 months of life in preterm infants (n = 27) compared with full-term babies (n = 13).
Results Immaturity affects the microbiota as indicated by a reduced percentage of the family Bacteroidaceae during the first months of life and by a higher initial percentage of Lactobacillaceae in preterm infants compared with full term infants. Perinatal antibiotics, including intrapartum antimicrobial prophylaxis, affects the gut microbiota, as indicated by increased Enterobacteriaceae family organisms in the infants.

Human gut microbiome

Human gut microbiome

Conclusions Prematurity and perinatal antibiotic administration strongly affect the initial establishment of microbiota with potential consequences for later health.

Ischemia and necrotizing enterocolitis: where, when, and how

Philip T. Nowicki
Seminars in Pediatric Surgery (2005) 14, 152-158
http://dx.doi.org:/10.1053/j.sempedsurg.2005.05.003

While it is accepted that ischemia contributes to the pathogenesis of necrotizing enterocolitis (NEC), three important questions regarding this role subsist. First, where within the intestinal circulation does the vascular pathophysiology occur? It is most likely that this event begins within the intramural microcirculation, particularly the small arteries that pierce the gut wall and the submucosal arteriolar plexus insofar as these represent the principal sites of resistance regulation in the gut. Mucosal damage might also disrupt the integrity or function of downstream villous arterioles leading to damage thereto; thereafter, noxious stimuli might ascend into the submucosal vessels via downstream venules and lymphatics. Second, when during the course of pathogenesis does ischemia occur? Ischemia is unlikely to the sole initiating factor of NEC; instead, it is more likely that ischemia is triggered by other events, such as inflammation at the mucosal surface. In this context, it is likely that ischemia plays a secondary, albeit critical role in disease extension. Third, how does the ischemia occur? Regulation of vascular resistance within newborn intestine is principally determined by a balance between the endothelial production of the vasoconstrictor peptide endothelin-1 (ET-1) and endothelial production of the vasodilator free radical nitric oxide (NO). Under normal conditions, the balance heavily favors NO-induced vasodilation, leading to a low resting resistance and high rate of flow. However, factors that disrupt endothelial cell function, eg, ischemia-reperfusion, sustained low-flow perfusion, or proinflammatory mediators, alter the ET-1:NO balance in favor of constriction. The unique ET-1–NO interaction thereafter might facilitate rapid extension of this constriction, generating a viscous cascade wherein ischemia rapidly extends into larger portions of the intestine.

Schematic representation of the intestinal microcirculation

Schematic representation of the intestinal microcirculation

Schematic representation of the intestinal microcirculation. Small mesenteric arteries pierce the muscularis layers and terminate in the submucosa where they give rise to 1A (1st order) arterioles. 2A (2nd order) arterioles arise from the 1A. Although not shown here, these 2A arterioles connect merge with several 1A arterioles, thus generating an arteriolar plexus, or manifold that serves to pressurize the terminal downstream microvasculature. 3A (3rd order) arterioles arise from the 2A and proceed to the mucosa, giving off a 4A branch just before descent into the mucosa. This 4A vessel travels to the muscularis layers. Each 3A vessel becomes the single arteriole perfusing each villus.

Collectively, these studies indicate that disruption of endothelial cell function has the potential to disrupt the normal balance between NO and ET-1 within the newborn intestinal circulation, and that such an event can generate significant ischemia. In this context, it is important to note that NO and ET-1 each regulate the expression and activity of the other. An increased [NO] within the microvascular environment reduces ET-1 expression and compromises ligand binding to the ETA receptor (thus decreasing its contractile efficacy), while ET-1 compromises eNOS expression. Thus, factors that upset the balance between NO and ET-1 will have an immediate and direct effect on vascular tone, but also exert an additional indirect effect by extenuating the disruption of balance between these two factors.

It is not difficult to construct a hypothesis that links the perturbations of I/R and sustained low-flow perfusion with an initial inflammatory insult. Initiation of an inflammatory process at the mucosal–luminal interface could have a direct impact on villus and mucosal 3A arterioles, damaging arteriolar integrity and disrupting villus hemodynamics. Ascent of proinflammatory mediators to the submucosal 1A–2A arteriolar plexus could occur via draining venules and lymphatics, generating damage to vascular effector systems therein; these mediators might include cytokines and platelet activating factor, as these elements have been recovered from human infants with NEC. This event, coupled with a generalized loss of 3A flow throughout a large portion of the mucosal surface, could compromise flow rate within the submucosal arteriolar plexus.

Necrotizing enterocolitis: An update

Loren Berman, R. Lawrence Moss
Seminars in Fetal & Neonatal Medicine 16 (2011) 145e150
http://dx.doi.org:/10.1016/j.siny.2011.02.002

Necrotizing enterocolitis (NEC) is a leading cause of death among patients in the neonatal intensive care unit, carrying a mortality rate of 15e30%. Its pathogenesis is multifactorial and involves an over reactive response of the immune system to an insult. This leads to increased intestinal permeability, bacterial translocation, and sepsis. There are many inflammatory mediators involved in this process, but thus far none has been shown to be a suitable target for preventive or therapeutic measures. NEC usually occurs in the second week of life after the initiation of enteral feeds, and the diagnosis is made based on physical examination findings, laboratory studies, and abdominal radiographs. Neonates with NEC are followed with serial abdominal examinations and radiographs, and may require surgery or primary peritoneal drainage for perforation or necrosis. Many survivors are plagued with long term complications including short bowel syndrome, abnormal growth, and neurodevelopmental delay. Several evidence-based strategies exist that may decrease the incidence of NEC including promotion of human breast milk feeding, careful feeding advancement, and prophylactic probiotic administration in at-risk patients. Prevention is likely to have the greatest impact on decreasing mortality and morbidity related to NEC, as little progress has been made with regard to improving outcomes for neonates once the disease process is underway.

Immune Deficiencies

Primary immunodeficiencies: A rapidly evolving story

Nima Parvaneh, Jean-Laurent Casanova,  LD Notarangelo, ME Conley
J Allergy Clin Immunol 2013;131:314-23.
http://dx.doi.org/10.1016/j.jaci.2012.11.051

The characterization of primary immunodeficiencies (PIDs) in human subjects is crucial for a better understanding of the biology of the immune response. New achievements in this field have been possible in light of collaborative studies; attention paid to new phenotypes, infectious and otherwise; improved immunologic techniques; and use of exome sequencing technology. The International Union of Immunological Societies Expert Committee on PIDs recently reported on the updated classification of PIDs. However, new PIDs are being discovered at an ever-increasing rate. A series of 19 novel primary defects of immunity that have been discovered after release of the International Union of Immunological Societies report are discussed here. These new findings highlight the molecular pathways that are associated with clinical phenotypes and suggest potential therapies for affected patients.

Combined Immunodeficiencies

  • T-cell receptor a gene mutation: T-cell receptor ab1 T-cell depletion

T cells comprise 2 distinct lineages that express either ab or gd T-cell receptor (TCR) complexes that perform different tasks in immune responses. During T-cell maturation, the precise order and efficacy of TCR gene rearrangements determine the fate of the cells. Productive β-chain gene rearrangement produces a pre-TCR on the cell surface in association with pre-Tα invariant peptide (β-selection). Pre-TCR signals promote α-chain recombination and transition to a double-positive stage (CD41CD81). This is the prerequisite for central tolerance achieved through positive and negative selection of thymocytes.

  • Ras homolog gene family member H deficiency: Loss of naive T cells and persistent human papilloma virus infections
  • MST1 deficiency: Loss of naive T cells

New insight into the role of MST1 as a critical regulator of T-cell homing and function was provided by the characterization of 8 patients from 4 unrelated families who had homozygous nonsense mutations in STK4, the gene encoding MST1. MST1 was originally identified as an ubiquitously expressed kinase with structural homology to yeast Ste. MST1 is the mammalian homolog of the Drosophila Hippo protein, controlling cell growth, apoptosis, and tumorigenesis. It has both proapoptotic and antiapoptotic functions.

  • Lymphocyte-specific protein tyrosine kinase deficiency: T-cell deficiency with CD41 lymphopenia

Defects in pre-TCR– and TCR-mediated signaling lead to aberrant T-cell development and function (Fig). One of the earliest biochemical events occurring after engagement of the (pre)-TCR is the activation of lymphocyte-specific protein tyrosine kinase (LCK), a member of the SRC family of protein tyrosine kinases. This kinase then phosphorylates immunoreceptor tyrosine-based activation motifs of intracellular domains of CD3 subunits. Phosphorylated immunoreceptor tyrosine-based activation motifs recruit z-chain associated protein kinase of 70 kDa, which, after being phosphorylated by LCK, is responsible for activation of critical downstream events. Major consequences include activation of the membrane-associated enzyme phospholipase Cg1, activation of the mitogen-activated protein kinase, nuclear translocation of nuclear factor kB (NFkB), and Ca21/Mg21 mobilization. Through these pathways, LCK controls T-cell development and activation. In mice lacking LCK, T-cell development in the thymus is profoundly blocked at an early double-negative stage.

TCR signaling

TCR signaling

TCR signaling. Multiple signal transduction pathways are stimulated through the TCR. These pathways collectively activate transcription factors that organize T-cell survival, proliferation, differentiation, homeostasis, and migration. Mutant molecules in patients with TCR-related defects are indicated in red.

  • Uncoordinated 119 deficiency: Idiopathic CD41 lymphopenia

Idiopathic CD41 lymphopenia (ICL) is a very heterogeneous clinical entity that is defined, by default, by persistent CD41 T-cell lymphopenia (<300 cells/mL or <20% of total T cells) in the absence of HIV infection or any other known cause of immunodeficiency.

Well-Defined Syndromes with Immunodeficiency

  • Wiskott-Aldrich syndrome protein–interacting protein deficiency: Wiskott-Aldrich syndrome-like phenotype

In hematopoietic cells Wiskott-Aldrich syndrome protein (WASP) is stabilized through forming a complex with WASP interacting protein (WIP).

  • Phospholipase Cg2 gain-of-function mutations: Cold urticaria, immunodeficiency, and autoimmunity/autoinflammatory

This is a unique phenotype, sharing features of antibody deficiency, autoinflammatory diseases, and immune dysregulatory disorders, making its classification difficult. Two recent studies validated the pleiotropy of genetic alterations in the same gene.

Predominantly Antibody Defects

  • Defect in the p85a subunit of phosphoinositide 3-kinase: Agammaglobulinemia and absent B cells
  • CD21 deficiency: Hypogammaglobulinemia
  • LPS-responsive beige-like anchor deficiency:
  • Hypogammaglobulinemia with autoimmunity and

early colitis

Defects Of Immune Dysregulation

  • Pallidin deficiency: Hermansky-Pudlak syndrome type 9
  • CD27 deficiency: Immune dysregulation and
  • persistent EBV infection

Congenital Defects Of Phagocyte Number, Function, Or Both

  • Interferon-stimulated gene 15 deficiency: Mendelian susceptibility to mycobacterial diseases

Defects In Innate Immunity

  • NKX2-5 deficiency: Isolated congenital asplenia
  • Toll/IL-1 receptor domain–containing adaptor inducing IFN-b and TANK-binding kinase 1 deficiencies: Herpes simplex encephalitis
  • Minichromosome maintenance complex component 4 deficiency: NK cell deficiency associated with growth retardation and adrenal insufficiency

Autoinflammatory Disorders

  • A disintegrin and metalloproteinase 17 deficiency: Inflammatory skin and bowel disease

 

Cross-talk between monocyte.macrophage cells and T.NK lymphocytes

Cross-talk between monocyte.macrophage cells and T.NK lymphocytes

Cross-talk between monocyte/macrophage cells and T/NK lymphocytes. Genes in the IL-12/IFN-g pathway are particularly important for protection against mycobacterial disease. IRF8 is an IFN-g–inducible transcription factor required for the induction of various target genes, including IL-12. The NF-kB essential modulator (NEMO) mutations in the LZ domain impair CD40-NEMO–dependent pathways. Some gp91phox mutations specifically abolish the respiratory burst in monocyte-derived macrophages. ISG15 is secreted by neutrophils and potentiates IFN-g production by NK/T cells. Genetic defects that preclude monocyte development (eg, GATA2) can also predispose to mycobacterial infections (not shown). Mutant molecules in patients with unusual susceptibility to infection are indicated in red.

The field of PIDs is advancing at full speed in 2 directions. New genetic causes of known PIDs are being discovered (eg, CD21 and TRIF). Moreover, new phenotypes qualify as PIDs with the identification of a first genetic cause (eg, generalized pustular psoriasis). Recent findings contribute fundamental knowledge about immune system biology and its perturbation in disease. They are also of considerable clinical benefit for the patients and their families. A priority is to further translate these new discoveries into improved diagnostic methods and more effective therapeutic strategies, promoting the well-being of patients with PIDs.

Primary immunodeficiencies

Luigi D. Notarangelo
J Allergy Clin Immunol 2010; 125(2): S182-194
http://dx.doi.org:/10.1016/j.jaci.2009.07.053

In the last years, advances in molecular genetics and immunology have resulted in the identification of a growing number of genes causing primary immunodeficiencies (PIDs) in human subjects and a better understanding of the pathophysiology of these disorders. Characterization of the molecular mechanisms of PIDs has also facilitated the development of novel diagnostic assays based on analysis of the expression of the protein encoded by the PID-specific gene. Pilot newborn screening programs for the identification of infants with severe combined immunodeficiency have been initiated. Finally, significant advances have been made in the treatment of PIDs based on the use of subcutaneous immunoglobulins, hematopoietic cell transplantation from unrelated donors and cord blood, and gene therapy. In this review we will discuss the pathogenesis, diagnosis, and treatment of PIDs, with special attention to recent advances in the field.

 

 

Read Full Post »

« Newer Posts - Older Posts »