Funding, Deals & Partnerships: BIOLOGICS & MEDICAL DEVICES; BioMed e-Series; Medicine and Life Sciences Scientific Journal – http://PharmaceuticalIntelligence.com
Reporter: Danielle Smolyar, Research Assistant 3 – Text Analysis for 2.0 LPBI Group’s TNS #1 – 2020/2021 Academic Internship in Medical Text Analysis (MTA)
Recently, researchers at Mount Sinai were able to develop a therapeutic agent that shows high levels of effectiveness in Vitro disrupting a biological pathway that allow cancer to survive. This finding is according to a paper which was published in Cancer Discovery, which is a Journal of the American Association of cancer research in July 2021.
The therapy in which they focus on is a molecule named MS21, which causes the degradation of AKT which is an enzyme that is very active and present in cancers. In this study there was much evidence that pharmacological degradation of AKT is a feasible treatment for cancer’s which have a mutation in certain genes.
AKT is a cancer gene that encodes an enzyme that is abnormally activated in cancer cells to stimulate tumor growth. The degradation of AKT reverses all these processes which ultimately inhibits further tumor growth.
“Our study lays a solid foundation for the clinical development of an AKT degrader for the treatment of human cancers with certain gene mutations,” said Ramon Parsons, MD, Ph.D., Director of The Tisch Cancer Institute and Ward-Coleman Chair in Cancer Research and Chair of Oncological Sciences at the Icahn School of Medicine at Mount Sinai. “Examination of 44,000 human cancers identified that 19 percent of tumors have at least one of these mutations, suggesting that a large population of cancer patients could benefit from therapy with an AKT degrader such as MS21.”
MS21 was tested and human cancer derived cell lines, is used in Laboratories as a model to study the efficacy of different cancer therapies.
At Mount Sinai they were looking to develop MS21 with an industry partner in order to open clinical trials for patients.
“Translating these findings into effective cancer therapies for patients is a high priority because the mutations and the resulting cancer-driving pathways that we lay out in this study are arguably the most commonly activated pathways in human cancer, but this effort has proven to be particularly challenging,” said Jian Jin, Ph.D., Mount Sinai Professor in Therapeutics Discovery and Director of the Mount Sinai Center for Therapeutics Discovery at Icahn Mount Sinai. “We look forward to an opportunity to develop this molecule into a therapy that is ready to be studied in clinical trials.”
Advancing cancer precision medicine by creating a better toolbox for cancer therapy
Jian Jin1,2,3,4,5*, Arvin C. Dar1,2,3,4, Deborah Doroshow1
A
mong approximately 20,000 proteins in the human proteome, 627 have been identified by cancer-dependency studies as priority cancer targets, which are functionally important for various cancers. Of these 600-plus priority targets, 232 are enzymes and 395 are nonenzyme proteins (1). Tremendous progress has been made over the past several decades in targeting enzymes, in particular kinas-es, which have suitable binding pockets that can be occupied by small-molecule inhibitors, leading to U.S. Food and Drug Administration (FDA) approvals of many small-molecule drugs as targeted anticancer thera-
1Tisch Cancer Institute; 2Department of Oncological Sciences; 3Department of Pharmacological Sciences; 4Mount Sinai Center for Therapeutics Discovery; 5Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
pies. However, most of the 395 nonenzyme protein targets, including transcription factors (TFs), do not have suitable binding pockets that can be effectively targeted by small molecules. These targets have consequently been considered undruggable; however, new cutting-edge approaches and technologies have recently been developed to target some of these “un-druggable” proteins in order to advance precision oncology.
TPD, a promising approach to precision cancer therapeutics
Targeted protein degradation (TPD) refers to the process of chemically eliminating proteins of interest (POIs) by utilizing small molecules, which are broadly divided into two types of modalities: PROteolysis Targeting Chimeras (PROTACs) and molecular glues (2). PROTACs are het-erobifunctional small molecules that contain two moieties: one binding the POI, linked to another binding an ubiquitin E3 ligase. The induced proximity between the POI and ubiquitination machinery leads to selective polyubiquitylation of the POI and its subsequent degradation by the ubiquitin–proteasome system (UPS). Molecular glues are monovalent small molecules, which, when built for TPD, directly induce interactions between the POI and an E3 ligase, also resulting in polyubiquitylation and subsequent degradation of the POI by the UPS. One of the biggest potential advantages of these therapeutic modalities over traditional inhibitors is that PROTACs and molecular glues can target undruggable proteins. Explosive growth has been seen in the TPD field over recent years (2, 3). Here, we highlight several recent advancements.
TF-PROTAC, a novel platform for targeting undruggable
tumorigenic TFs
Many undruggable TFs are tumorigenic. To target them, TF-PROTAC was developed (4), which exploits the fact that TFs bind DNA in a sequence-specific manner. TF-PROTAC was created to selectively bind a TF and E3 ligase simultaneously, by conjugating a DNA oligonucleotide specific for the TF of interest to a selective E3 ligase ligand. As stated earlier, this simultaneous binding and induced proximity leads to selective polyubiquitination of the TF and its subsequent degradation by the UPS. TF-PROTAC is a cutting-edge technology that could potentially provide a universal strategy for targeting most undruggable tumorigenic TFs.
Development of novel PROTAC degraders
WDR5, an important scaffolding protein, not an enzyme, is essential for sustaining tumorigenesis in multiple cancers, including MLL-rearranged (MLL-r) leukemia. However, small-molecule inhibitors that block the pro-tein–protein interaction (PPI) between WDR5 and its binding partners exhibit very modest cancer cell–killing effects, likely due to the confounding fact that these PPI inhibitors target only some—but not all—of WDR5’s on-cogenic functions. To address this shortcoming, a novel WDR5 PROTAC, MS67, was recently created using a powerful approach that effectively eliminates the protein and thereby all WDR5 functions via ternary complex structure-based design (Figure 1) (5). MS67 is a highly effective WDR5 degrader that potently and selectively degrades WDR5 and effectively suppresses the proliferation of tumor cells both in vitro and in vivo. This study provides strong evidence that pharmacological degradation of WDR5 as a novel therapeutic strategy is superior to WDR5 PPI inhibition for treating WDR5-dependent cancers.
EZH2 is an oncogenic methyltransferase that catalyzes histone H3 lysine 27 trimethylation, mediating gene repression. In addition to this canonical function, EZH2 has numerous noncanonical tumorigenic functions. EZH2 enzymatic inhibitors, however, are generally ineffective in
suppressing tumor growth in triple-negative breast cancer (TNBC) and MLL-r leukemia models and fail to phenocopy antitumor effects induced by EZH2 knockdown strategies. To target both canonical and noncanon-ical oncogenic functions of EZH2, several novel EZH2 degraders were recently developed, including MS1943, a hydrophobic tag–based EZH2 degrader (6), and MS177, an EZH2 PROTAC (7). MS1943 and MS177 effectively degrade EZH2 and suppress in vitro and in vivo growth in TNBC and MLL-r leukemia, respectively, suggesting that EZH2 degraders could provide a novel and effective therapeutic strategy for EZH2-dependent tumors.
MS21, a novel AKT PROTAC degrader, was developed to target activated AKT, the central node of the PI3K–AKT–mTOR signaling pathway (8). MS21 effectively suppresses the proliferation of PI3K–PTEN pathway-mutant cancers with wild-type KRAS and BRAF, which represent a large percentage of all human cancers. Another recent technology that expands the bifunctional toolbox for TPD is the demonstration that the E3 ligase KEAP1 can be leveraged for PROTAC development using a selective KEAP1 ligand (9). Overall, tremendous progress has been made in discovering novel degraders, some of which have advanced to clinical development as targeted therapies (2, 3).
Novel approaches to selective TPD in cancer cells
To minimize uncontrolled protein degradation in normal tissues, which may cause potential toxicity, a new technology was developed that incorporates a light-inducible switch, termed “opto-PROTAC” (10). This switch serves as a caging group that renders opto-PROTAC inactive in all cells in the absence of ultraviolet (UV) light. Upon UV irradiation, however, the caging group is removed, resulting in the release of the active degrader and spatiotemporal control of TPD in cancer cells. Another strategy to achieve selective TPD in cancer over normal cells is to cage degraders with a folate group (11, 12). Folate-caged degraders are inert and selectively concentrated within cancer cells, which overexpress folate receptors compared to normal cells. The caging group is subsequently removed inside tumor cells, releasing active degraders and achieving selective TPD in these cells. These novel approaches potentially enable degraders to be precision cancer medicines.
11
Frontiers of Medical Research: Cancer
Trametiglue, a novel and atypical molecular glue
The RAS–RAF–MEK–ERK signaling pathway, one of the most frequently mutated pathways in cancer, has been intensively targeted. Several drugs, such as the KRAS G12C inhibitor sotorasib and the MEK inhibitor trametinib, have been approved by the FDA. A significant advancement in this area is the discovery that trametinib unexpectedly binds a pseudokinase scaffold termed “KSR” in addition to MEK through interfacial contacts (13). Based on this structural and mechanistic insight, tra-metiglue, an analog of trametinib, was created as a novel molecular glue to limit adaptive resistance to MEK inhibition by enhancing interfacial binding between MEK, KSR, and the related homolog RAF. This study provides a strong foundation for developing next-generation drugs that target the RAS pathway.
TF-DUBTAC, a novel technology to stabilize undruggable tumor-suppressive TFs
Complementary to degrading tumorigenic TFs, stabilizing tumor-suppressive TFs could provide another effective approach for treating cancer. While most tumor-suppressive TFs are undruggable, TF-DUBTAC was recently developed as a generalizable platform to stabilize tumor-suppressive TFs (14). Deubiquitinase-targeting chimeras (DUBTACs) are heterobifunctional small molecules with a deubiquitinase (DUB) ligand linked to a POI ligand, which stabilize POIs by harnessing the deubiq-uitination machinery (15). Similar to TF-PROTAC, TF-DUBTAC exploits the fact that most TFs bind specific DNA sequences. TF-DUBTAC links a DNA oligonucleotide specific to a tumor-suppressive TF with a selective DUB ligand, resulting in simultaneous binding of the TF and DUB. The induced proximity between the TF and DUB leads to selective deubiquiti-
Putting a bull’s-eye on cancer’s back
Scientists are aiming the immune systems’ “troops” directly at tumors to better treat cancer
Joshua D. Brody, Brian D. Brown
I
mmunotherapy has transformed the treatment of several types of cancers. In particular, immune checkpoint blockade (ICB), which reinvigorates killer T cells, has helped extend the lives of many patients with advanced-stage lung, bladder, kidney, or skin cancers. Unfortunately, ~80% of patients do not respond to current immunotherapies or even-tually relapse. Emerging data indicate that one of the most profound ways cancers resist immunotherapy is by keeping killer T cells out of the tumor and putting other immune cells in a suppressed state (1). This understanding is giving rise to a new frontier in immunotherapy that is using synthetic biology and other approaches to reprogram the tumor from immune “cold” to immune “hot,” so T cells can be recruited to the tumor, and enter, target, and destroy the cancer cells (2) (Figure 1).
Cancers protect themselves by keeping out immune cells
Cancers grow in tissues like foreign invaders. Though they start from healthy cells, mutations turn cells malignant and allow them to grow unchecked. T cells can kill malignant cells that express mutated proteins, but cancers employ strategies to fend off the T cells. One way they do this is
12
nation of the TF and its stabilization. As an exciting new technology, TF-DUBTAC provides a potential general strategy to stabilize most undrugga-ble tumor-suppressive TFs for treating cancer.
Future outlook
The breathtaking pace we are seeing in the development of innovative approaches and technologies for advancing cancer therapies is only expected to accelerate. The promising clinical results achieved by PROTACs with established targets are particularly encouraging and pave the way for development of PROTACs for newer and more innovative targets. These groundbreaking discoveries have now put opportunities to fully realize cancer precision medicine within our reach.
References
F. M. Behan et al., Nature 568, 511–516 (2019).
B. Dale et al., Nat. Rev. Cancer 21, 638–654 (2021).
A. Mullard, Nat. Rev. Drug Discov. 20, 247–250 (2021).
J. Liu et al., J. Am. Chem. Soc. 143, 8902–8910 (2021).
X. Yu et al., Sci. Transl. Med. 13, eabj1578 (2021).
A. Ma et al., Nat. Chem. Biol. 16, 214–222 (2020).
J. Wang et al., Nat. Cell Biol. 24, 384–399 (2022).
J. Xu et al., Cancer Discov. 11, 3064–3089 (2021).
J. Wei et al., J. Am. Chem. Soc. 143, 15073–15083 (2021).
J. Liu et al., Sci. Adv. 6, eaay5154 (2020).
J. Liu et al., J. Am. Chem. Soc. 143, 7380–7387 (2021).
H. Chen et al., J. Med. Chem. 64, 12273–12285 (2021).
Z. M. Khan et al., Nature 588, 509–514 (2020).
J. Liu et al., J. Am. Chem. Soc. 144, 12934–12941 (2022).
N. J. Henning et al., Nat. Chem. Biol. 18, 412–421 (2022
Other related articles published on this Open Access Online Scientific Journal include the following:
Machine Learning (ML) in cancer prognosis prediction helps the researcher to identify multiple known as well as candidate cancer diver genes
DISCUSSION – Genomics-driven personalized medicine for Pancreatic Cancer, Volume 2 (Volume Two: Latest in Genomics Methodologies for Therapeutics: Gene Editing, NGS and BioInformatics, Simulations and the Genome Ontology), Part 1: Next Generation Sequencing (NGS)
DISCUSSION – Genomics-driven personalized medicine for Pancreatic Cancer
Reporter: Aviva Lev-Ari, PhD, RN
[bold face added, ALA]
Integrated Patient-Derived Models Delineate Individualized Therapeutic Vulnerabilities of Pancreatic Cancer –>>> Personalized Tumor Models Could Help Identify Combination Therapies for Hard-to-Treat Cancers
PDAC has a particularly poor prognosis, and even with new targeted therapies and chemotherapy, the survival is poor. Here, we show that patient-derived models can be developed and used to investigate therapeutic sensitivities determined by genetic features of the disease and to identify empirical therapeutic vulnerabilities. These data reveal several key points that are of prime relevance to pancreatic cancer and tumor biology in general.
The Challenges of Using Genetic Analysis to Inform Treatment in PDAC
Precision oncology is dependent on the existence of known vulnerabilities encoded by high-potency genetic events and drugs capable of exploiting these vulnerabilities. At present, the repertoire of actionable genetic events in PDAC is limited.
Rare BRAF V600E mutations are identified in PDAC and could represent the basis for targeted inhibition, as our group and others have previously published (Collisson et al., 2012; Witkiewicz et al., 2015).
Similarly,
germline BRCA deficiency is the basis for ongoing poly(ADP-ribose) polymerase (PARP) inhibitor clinical trials (Lowery et al., 2011).
As shown here, out of 28 cases, only one genetic event was identified that yielded sensitivity to a therapeutic strategy. In this case, existence of the matched model allowed us to confirm the biological relevance of the
STAG2 mutation by showing sensitivity of the model to a DNA cross-linking agent.
Therefore, annotated patient-derived models provide a substrate upon which to functionally dissect the significance of novel and potentially actionable genetic events that occur within a tumor.
Another challenge of genomics-driven personalized medicine is
assessing the effect of specific molecular aberrations on therapeutic response in the context of complex genetic changes present in individual tumors.
KRAS has been proposed to modify therapeutic dependency to EZH2 inhibitors (Kim et al., 2015), and in the models tested, responses to this class of drugs were not uniformly present in cases harboring mutations in chromatin-remodeling genes.
This finding suggests that, although tumors acquire genetic alterations in specific genes, the implicated pathway may not be functionally inactive or therapeutically actionable. Therefore, annotated patient derived models provide a unique test bed for interrogating specific therapeutic dependencies in a genetically tractable system.
Empirical Definition of Therapeutic Sensitivities and Clinical Relevance
Cell lines offer the advantage of the ability to conduct high throughput approaches to interrogate many therapeutic agents. A large number of failed clinical trials have demonstrated the difficulty in treating PDAC. Based on the data herein, the paucity of clinical success is, most probably, due to the diverse therapeutic sensitivity of individual PDAC cases, suggesting that, with an unselected patient population, it will be veritably impossible to demonstrate clinical benefit. Additionally,
very few models exhibited an exceptional response to single agents across the breadth of a library encompassing 305 agents.
We could identify only one tumor that was particularly sensitive to MEK inhibition and another model that was sensitive to
EGFR and
tyrosine kinase inhibitors.
In contrast to the limited activity of single agents, combination screens yielded responses at low-dose concentrations in the majority of models. Specific combinations were effective across several models, indicating that, by potentially screening more models, therapeutic sensitivity clades of PDAC will emerge. In the pharmacological screens performed in this study,
MEK inhibition, coupled with MTOR, docetaxel, or tyrosine kinase inhibitors, was effective in _30% of models tested.
Resistance to MEK inhibitors occurs through several mechanisms, including
Upregulation of oncogenic bypass signaling pathways such as AKT, tyrosine kinase, or MTOR (mammalian target of rapamycin) signaling.
In the clinic, the MEK and MTOR inhibitors (e.g., NCT02583542) are being tested. An intriguing finding from the drug screen was
sensitivity of a subset of models to combined MEK and docetaxel inhibition. This combination has been observed to synergistically enhance apoptosis and inhibit tumor growth in human xenograft tumor models (Balko et al., 2012; McDaid et al., 2005) and is currently being tested in a phase III study in patients with KRAS-mutated, advanced non-small-cell lung adenocarcinoma (Ja¨ nne et al., 2016).
Interestingly, in the models tested herein, there was limited sensitivity imparted through
the combination of gemcitabine and MEK inhibition.
This potentially explains why the combination of MEK inhibitor and gemcitabine tested in the clinic did not show improved efficacy over gemcitabine alone (Infante et al., 2014).
Another promising strategy that emerged from this study involves using
CHK or BCL2 inhibitors as agents that drive enhanced sensitivity to chemotherapy.
Together, the data suggest that the majority of PDAC tumors have intrinsic therapeutic sensitivities, but the challenge is to prospectively identify effective treatment.
Patient-Derived Model-Based Approach to Precision Medicine
This study supports a path for guiding patient treatment based on the integration of genetic and empirically determined sensitivities of the patient’s tumor (Figure S7). In reference to defined genetic susceptibilities, the models provide a means to interrogate the voracity of specific drug targets. Parallel unbiased screening enables the discovery of sensitivities that could be exploited in the clinic. The model-guided treatment must be optimized, allowing for the generation of data in a time frame compatible with clinical decision making and appropriate validation.
In the present study, the majority of models were developed, cell lines were drug screened, and select hits were validated in PDX models within a 10- to 12-month window (Figure S7). This chronology would allow time to inform frontline therapy for recurrent disease for most patients who were surgically resected and treated with a standard of care where the median time to recurrence is approximately 14 months (Saif, 2013).
Although most models were generated from surgically resected specimens, two of the models (EMC3226 and EMC62) were established from primary tumor biopsies, indicating that this approach could be used with only a limited amount of tumor tissue available.
In the context of inoperable pancreatic cancer, application of data from a cell-line screen without in vivo validation in PDX would permit the generation of sensitivity data in the time frame compatible with treatment.
[We] acknowledge that model-guided treatment is also not without significant logistical hurdles, including the availability of drugs for patient treatment, clinically relevant time frames, patient-performance status, toxicity of combination regiments, and quality metrics related to model development and therapeutic response evaluation.
Additionally, it will be very important to monitor ex vivo genetic and phenotypic divergence with passage and try to understand the features of tumor heterogeneity that could undermine the efficacy of using models to direct treatment. As shown here, drug sensitivities remained stable with passage in cell culture and, importantly, were confirmed in PDX models, suggesting that the dominant genetic drivers and related therapeutic sensitivities are conserved.
In spite of these challenges, progressively more effort is going into the development of patient-derived models for guidance of disease treatment (Aparicio et al., 2015; Boj et al., 2015; Crystal et al., 2014; van de Wetering et al., 2015).
Several ongoing trials use PDX models to direct a limited repertoire of agents (e.g., NCT02312245, NCT02720796, and ERCAVATAR2015). Given the experience here, PDAC cell lines would provide the opportunity to rapidly interrogate a larger portfolio of combinations that could be used to guide patient care and provide a novel approach to precision medicine.
Validation of this approach would require the establishment of challenging multi-arm or N-of-1 clinical trials. However, considering the dire outcome for PDAC patients and the long-lasting difficulty in developing effective treatments, this non-canonical approach might be particularly impactful in pancreatic cancer.
Whereas the functional link between HDL and eNOS has been appreciated only recently, the relationship between HDL and endothelium-dependent vasodilation has been known for some time. In studies of coronary vasomotor responses to acetylcholine, it was noted in 1994 that patients with elevated HDL have greater vasodilator and attenuated vasoconstrictor responses (Zeiher et al., 1994).
Studies of flow-mediated vasodilation of the brachial artery have also shown that HDL cholesterol is an independent predictor of endothelial function (Li et al., 2000).
Induction of NO Production and Stimulation of eNOS
Mechanism of Action (MOA) for Nitric Oxide (NO) and endothelial Nitric Oxide Syntase (eNOS) are described in George T. and P. Ramwell, (2004). Nitric Oxide, Donors, & Inhibitors. Chapter 19 in Katzung, BG., Basic & Clinical Pharmacology. McGraw-Hill, 9th Edition, pp. 313 – 318
The direct, short-term impact of HDL on endothelial function also has recently been investigated in humans. One particularly elegant study recently evaluated forearm blood flow responses in individuals who are heterozygous for a loss-of-function mutation in the ATP-binding cassette transporter 1 (ABCA1) gene. Compared with controls, ABCA1 heterozygotes (six men and three women) had HDL levels that were decreased by 60%, their blood flow responses to endothelium-dependent vasodilators were blunted, and endothelium-independent responses were unaltered. After a 4-hour infusion of apoAI/phosphatidylcholine disks, their HDL level increased threefold and endothelium-dependent vasomotor responses were fully restored (Bisoendial et al., 2003). It has also been observed that endothelial function is normalized in hypercholesterolemic men with normal HDL levels shortly following the administration of apoAI/phosphatidylcholine particles (Spieker et al., 2002).
In addition to the modulation of NO production by signaling events that rapidly dictate the level of enzymatic activity, important control of eNOS involves changes in the abundance of the enzyme. In a clinical trial by the Karas laboratory of niacin therapy in patients with low HDL levels (nine males and two females), flow-mediated dilation of the brachial artery was improved in association with a rise in HDL of 33% over 3 months (Kuvin et al., 2002).
They also demonstrated that eNOS expression in cultured human endothelial cells is increased by HDL exposure for 24 hours. They further showed that the increase in eNOS is related to an increase in the half-life of the protein, and that this is mediated by PI3K–Akt kinase and MAPK (Ramet et al., 2003).
It describes the mechanism of action for HDL enhancement of NO production by eNOS in vascular endothelium.
(a) HDL causes membrane-initiated signaling, which stimulates eNOS activity. The eNOS protein is localized in cholesterol-enriched (orange circles) plasma membrane caveolae as a result of the myristoylation and palmitoylation of the protein. Binding of HDL to SR-BI via apoAI causes rapid activation of the nonreceptor tyrosine kinase src, leading to PI3K activation and downstream activation of Akt kinase and MAPK. Akt enhances eNOS activity by phosphorylation, and independent MAPK-mediated processes are additionally required (Duarte, et al., 1997). .Eur J Pharmacol, 338:25–33. HDL also causes an increase in intracellular Ca2+ concentration (intracellular Ca2+ store shown in blue; Ca2+ channel shown in pink), which enhances binding of calmodulin (CM) to eNOS. HDL-induced signaling is mediated at least partially by the HDL-associated lysophospholipids SPC, S1P, and LSF acting through the G protein–coupled lysophospholipid receptor S1P3. HDL-associated estradiol (E2) may also activate signaling by binding to plasma membrane–associated estrogen receptors (ERs), which are also G protein coupled. It remains to be determined if signaling events are also directly mediated by SR-BI (Yuhanna et al., 2001), (Nofer et al., 2004), (Gong et al., 2003), (Mineo et al., 2003).
(b) HDL regulates eNOS abundance and subcellular distribution. In addition to modulating the acute response, the activation of the PI3K–Akt kinase pathway and MAPK by HDL upregulates eNOS expression (open arrows). HDL also regulates the lipid environment in caveolae (dashed arrows). Oxidized LDL (OxLDL) can serve as a cholesterol acceptor (orange circles), thereby disrupting caveolae and eNOS function. However, in the presence of OxLDL, HDL maintains the total cholesterol content of caveolae by the provision of cholesterol ester (blue circles), resulting in preservation of the eNOS signaling module (Ramet et al., 2003), (Blair et al., 1999), (Uittenbogaard et al., 2000).
eNOS is not Activated by Nebivolol in Human Failing Myocardium.
Nebivolol is a highly selective beta(1)-adrenoceptor blocker with additional vasodilatory properties, which may be due to an endothelial-dependent beta(3)-adrenergic activation of the endothelial nitric oxide synthase (eNOS). beta(3)-adrenergic eNOS activation has been described in human myocardium and is increased in human heart failure. Therefore, this study investigated whether nebivolol may induce an eNOS activation in cardiac tissue. Immunohistochemical stainings were performed using specific antibodies against eNOS translocation and eNOS serine(1177) phosphorylation in rat isolated cardiomyocytes, human right atrial tissue (coronary bypass-operation), left ventricular non-failing (donor hearts) and failing myocardium after application of the beta-adrenoceptor blockers nebivolol, metoprolol and carvedilol, as well as after application of BRL 37344, a specific beta(3)-adrenoceptor agonist. BRL 37344 (10 muM) significantly increased eNOS activity in all investigated tissues (either via translocation or phosphorylation or both). None of the beta-blockers (each 10 muM), including nebivolol, increased either translocation or phosphorylation in any of the investigated tissues. In human failing myocardium, nebivolol (10 muM) decreased eNOS activity. In conclusion, nebivolol shows a tissue-specific eNOS activation. Nebivolol does not activate the endothelial eNOS in end-stage human heart failure and may thus reduce inhibitory effects of NO on myocardial contractility and on oxidative stress formation. This mode of action may be of advantage when treating heart failure patients.
Brixius K, Song Q, Malick A, Boelck B, Addicks K, Bloch W, Mehlhorn U, Schwinger R, (2006). eNOS is not activated by nebivolol in human failing myocardium.
Brixius K, Song Q, Malick A, Boelck B, Addicks K, Bloch W, Mehlhorn U, Schwinger R, (2006). eNOS is not activated by nebivolol in human failing myocardium.
In most solid cancers, cells harboring oncogenic mutations represent only a sub-fraction of the entire population. Within this sub-fraction the expression level of mutated proteins can vary significantly due to
cellular variability limiting the efficiency of targeted therapy.
To address the causes of the heterogeneity, we performed a systematic analysis of one of the most frequently mutated pathways in cancer cells, the phosphatidylinositol 3 kinase (PI3K) signaling pathway. Among others PI3K signaling is activated by the hepatocyte growth factor (HGF) that regulates
proliferation of hepatocytes during liver regeneration but
HGF-mediated responses of PI3K signaling were monitored both at the single cell and cell population level in primary mouse hepatocytes and in the hepatoma cell line Hepa1_6. Interestingly, we observed that the HGF-mediated AKT responses at the level of individual cells is rather heterogeneous. However, the overall average behavior of the single cells strongly resembled the dynamics of AKT activation
determined at the cell population level.
To gain insights into the molecular cause for the observed heterogeneous behavior of individual cells, we employed
dynamic mathematical modeling in a stochastic framework.
Our analysis demonstrated that intrinsic noise was not sufficient to explain the observed kinetic behavior, but rather
the importance of extrinsic noise has to be considered.
Thus, distinct from gene expression in the examined signaling pathway fluctuations of the reaction rates has only a minor impact whereas
variability in the concentration of the various signaling components even in a clonal cell population is a key determinant for the kinetic behavior.
English: Structure of the HGF protein. Based on PyMOL rendering of PDB 1bht. (Photo credit: Wikipedia)
Melanoma represents approximately 4% of human skin cancers, yet accounts for approximately 80% of deaths from cutaneous neoplasms. It remains one of the most common types of cancer among young adults. Melanoma is recognized as the most common fatal skin cancer with its incidence rising to 15 fold in the past 40 years in the United States. Melanoma develops from the malignant transformation of melanocytes, the pigment-producing cells that reside in the basal epidermal layer in human skin. (Greenlee RT, et al, Cancer J Clin. Jan-Feb 2001;51(1):15-36 ; Weinstock MA, et al, Med Health R I. Jul 2001;84(7):234-6). Classic clinical signs of melanoma include change in color, recent enlargement, nodularity, irregular borders, and bleeding. Cardinal signs of melanoma are sometimes referred to by the mnemonic ABCDEs (asymmetry, border irregularity, color, diameter, elevation) (Chudnovsky Y, et al. J Clin Invest, 1 April 2005; 115(4): 813–824).
Clinical characteristics
Melanoma primarily affects fair-haired and fair-skinned individuals, and those who burn easily or have a history of severe sunburn are at higher risk than their darkly pigmented, age-matched controls. The exact mechanism and wavelengths of UV light that are the most critical remain controversial, but both UV-A (wavelength 320–400 nm) and UV-B (290–320 nm) have been implicated (Jhappan C, et al, Oncogene, 19 May 2003;22(20):3099-112). Case-control studies have identified several risk factors in populations susceptible to developing melanoma. MacKie RM et al (1989) stated that the relative risk of cutaneous melanoma is estimated from the four strongest risk factors identified by conditional logistic regression. These factors are
total number of benign pigmented naevi above 2 mm diameter;
freckling tendency;
number of clinically atypical naevi (over 5 mm diameter and having an irregular edge, irregular pigmentation, or inflammation); and
As in many cancers, both genetic predisposition and exposure to environmental agents are risk factors for melanoma development. Many studies conducted over several decades on benign and malignant melanocytic lesions as well as melanoma cell lines have implicated numerous genes in melanoma development and progression.
Apart from the risk factors such as skin pigmentation, freckling, and so on, another significant risk factor is ‘strong family history of melanoma’. Older case-control studies of patients with familial atypical mole-melanoma (FAMM) syndrome suggested an elevated risk of ∼434-to 1000-fold over the general population (Greene MH, et al, Ann Intern Med, Apr 1985;102(4):458-65). A more recent meta-analysis of family history found that the presence of at least one first-degree relative with melanoma increases the risk by 2.24-fold (Gandini S, et al, Eur J Cancer, Sep 2005;41(14):2040-59). Genetic studies of melanoma-prone families have given important clues regarding melanoma susceptibility loci.
CDKN2A, the familial melanoma locus
CDKN2A is located at chromosome 9p21 and is composed of 4 exons (E) – 1α, 1β, 2, and 3. LOH or mutations at this locus cosegregated with melanoma susceptibility in familial melanoma kindred and 9p21 mutations have been observed in different cancer cell lines. The locus encodes two tumor suppressors via alternate reading frames, INK4 (p16INK4a) and ARF (p14ARF). INK4A and ARF encode alternative first exons, 1α and 1β respectively and different promoters. INK4A is translated from the splice product of E1α, E2, and E3, while ARF is translated from the splice product of E1β, E2, and E3. Second exons of the two proteins are shared and two translated proteins share no amino acid homology.
INK4A is the founding member of the INK4 (Inhibitor of cyclin-dependent kinase 4) family of proteins and inhibits the G1 cyclin-dependent kinases (CDKs) 4/6, which phosphorylate and inactivate the retinoblastoma protein (RB), thereby allowing for S-phase entry. Thus, loss of INK4K function promotes RB inactivation through hyperphosphorylation, resulting in unconstrained cell cycle progression.
ARF (Alternative Reading Frame) protein of the locus inhibits HDM2-mediated ubiquitination and subsequent degradation of p53. Thus, loss of ARF inactivates another tumor suppressor, p53. The loss of p53 impairs mechanisms that normally target genetically damaged cells for cell cycle arrest and/or apoptosis, which leads to proliferation of damaged cells. Loss of CDKN2A therefore contributes to tumorigenesis by disruption of both the pRB and p53 pathways.
Figure 1: Genetic encoding and mechanism of action of INK4A and ARF.
A genetic hallmark of melanoma is the presence of activating mutations in the oncogenes BRAF and NRAS, which are present in 70% and 15% of melanomas, respectively, and lead to constitutive activation of mitogen-activated protein kinase (MAPK) pathway signaling. However, molecules that inhibit MAPK pathway–associated kinases, like BRAF and MEK, have shown only limited efficacy in the treatment of metastatic melanoma. Thus, a deeper understanding of the cross talk between signaling networks and the complexity of melanoma progression should lead to more effective therapy.
NRAS mutations activate both effector pathways, Raf-MEK-ERK and PI3K-Akt in melanoma. The Raf-MEK-ERK pathway may also be activated via mutations in the BRAF gene. In a subset of melanomas, the ERK kinases have been shown to be constitutively active even in the absence of NRAS or BRAF mutations. The PI3K-Akt pathway may be activated through loss or mutation of the tumor suppressor gene PTEN, occurring in 30–50% of melanomas, or through gene amplification of the AKT3 isoform. Activation of ERK and/or Akt3 promotes the development of melanoma by various mechanisms, including stimulation of cell proliferation and enhanced resistance to apoptosis.
Figure 2: Schematic of the canonical Ras effector pathways Raf-MEK-ERK and PI3K-Akt in melanoma.
Curtin et al (2005) compared genome-wide alterations in the number of copies of DNA and mutational status of BRAF and NRAS in 126 melanomas from four groups in which the degree of exposure to ultraviolet light differs: 30 melanomas from skin with chronic sun-induced damage and 40 melanomas from skin without such damage; 36 melanomas from palms, soles, and subungual (acral) sites; and 20 mucosal melanomas. Significant differences were observed in number of copies of DNA and mutation frequencies in BRAF among the four groups of melanomas. Eighty-one percent of the melanomas on skin without sun-induced damaged had mutations in BRAF or NRAS. Melanomas with wild-type BRAF or NRAS frequently had increases in the number of copies of the genes for cyclin-dependent kinase 4 (CDK4) and cyclin D1 (CCND1), downstream components of the RAS-BRAF pathway. Thus, the genetic alterations identified in melanomas at different sites and with different levels of sun exposure indicate that there are distinct genetic pathways in the development of melanoma and implicate CDK4 and CCND1 as independent oncogenes in melanomas without mutations in BRAF or NRAS. (Curtin JA, et al, N Engl J Med, 17 Nov 2005;353(20):2135-47).
Genetic Heterogeneity of Melanoma
Melanoma exhibits molecular heterogeneity with markedly distinct biological and clinical behaviors. Lentigo maligna melanomas, for example, are indolent tumors that develop over decades on chronically sun-exposed area such as the face. Acral lentigenous melanoma, or the other hand, develops on sun-protected regions, tend to be more aggressive. Also, transcription profiling has provided distinct molecular subclasses of melanoma. It is also speculated that alterations at the DNA and RNA and the non-random nature of chromosomal aberrations may segregate melanoma tumors into subtypes with distinct clinical behaviors.
The melanoma gene atlas
Whole-genome screening technologies such as spectral karyotype analysis and array-CGH have identified many recurrent nonrandom chromosomal structural alterations, particularly in chromosomes 1, 6, 7, 9, 10, and 11 (Curtin JA, et al, N Engl J Med, 17 Nov 2005;353(20):2135-47); however, in most cases, no known or validated targets have been linked to these alterations.
In A systematic high-resolution genomic analysis of melanocytic genomes, array-CGH profiles of 120 melanocytic lesions, including 32 melanoma cell lines, 10 benign melanocytic nevi, and 78 melanomas (primary and metastatic) by Chin et al (2006) revealed a level of genomic complexity not previously appreciated. In total, 435 distinct copy number aberrations (CNAs) were defined among the metastatic lesions, including 163 recurrent, high-amplitude events. These include all previously described large and focal events (e.g., 1q gain, 6p gain/6q loss, 7 gain, 9p loss, and 10 loss). Genomic complexity observed in primary and benign nevi melanoma is significantly less than that observed in metastatic melanoma (Figure 3) (Chin L, et al, Genes Dev. 15 Aug 2006;20 (16):2149-2182).
Thus, genomic profiling of various melanoma progression types could reveal important information regarding genetic events those likely drive as metastasis and possibly, reveal provide cues regarding therapy targeted against melanoma.
Nitric Oxide and Sepsis, Hemodynamic Collapse, and the Search for Therapeutic Options
Curator, Reporter, EAW: Larry H Bernstein, MD, FCAP
This document explores the current understanding of sepsis as a cascade of events that involves the microcirculation unevenly because of a differential effect on the large and contiguous intestinal epithelium, secondary effects on cardiopulmonary blood flows and cardiac output, and the role of Nitric Oxide in the emergence of beneficial and potentially deleterious effects. This leads to a substantial body of work on therapeutic targets, either aimed at total inhibition or selective inhibition of NO synthase, and the special role of iNOS. This is another of a series of discussions on the metabolic and regulatory role of NO in health and disease.
Introduction
Antioxidants are essential, and are involved in several important biological processes such as immunity, protection against tissue damage, reproduction, growth and development. Antioxidants preserve adequate function of cells against homeostatic disturbances such as those caused by septic shock, aging and, in general, processes involving oxidative stress. This review focuses on the involvement of reactive oxygen and nitrogen species.
The presence of free radicals in biological materials was discovered about 50 years ago. Today, there is a large body of evidence indicating that patients in hospital intensive care units (ICUs) are exposed to excessive free radicals from drugs and other substances that alter cellular reduction -oxidation (redox) balance, and disrupt normal biological functions. However, low levels of free radicals are also vital for many cell signaling events and are essential for proper cell function.
Excess free radicals can result from a variety of conditions such as tissue damage and hypoxia (limiting oxygen levels), overexposure to environmental factors (tobacco smoke, ultraviolet radiation, and pollutants), a lack of antioxidants, or destruction of free radical scavengers. When the production of damaging free radicals exceeds the capacity of the body’s antioxidant defenses to detoxify them, a condition known as oxidative stress occurs.
A free radical can be described as any atom or a group of atoms or molecules in which there is at least one unpaired electron in the outermost shell . These free radicals are very reactive with adjacent molecules such as lipids, proteins, and carbohydrates and can cause cellular damage. Paradoxically, free radicals can also be produced by many cells as a protective mechanism, for example neutrophils produce free radicals to attack and destroy pathogens, while the liver uses free radicals for detoxification. However, the presence of free radicals within the body can also have a significant role in the development and progression of many disease processes for example heart disease, hypertension, cerebrovascular accidents, and diabetic complications. Any free radical involving O2 is referred to as a reactive oxygen species (ROS).
Normal cellular metabolism involves the production of ROS, and in humans, superoxide (O2 -) is the most commonly produced free radical. Phagocytic cells such as macrophages and neutrophils are prominent sources of O2 -. During an inflammatory response, these cells generate free radicals that attack invading pathogens such as bacteria and, because of this, the production of O2- by activated phagocytic cells in response to inflammation is one of the most studied free radical producing systems. The majority of the H2O2 is broken down to O2 and water by the antioxidant enzyme catalase. In addition to catalase, glutathione peroxidase can also break down H2O2 and also any peroxides that form on lipids within the body. When O2 – reacts with nitric oxide (NO), the toxic product peroxynitrite (ONOO-) is formed.
Cellular ROS originate from O2- generated as a by-product of oxidative phosphorylation (mitochondrial respiration), they differ in their mechanism of production, necessary cofactors, diffusion range, hydrophobicity, biological targets, detoxification pathways and breakdown products. O-2 damaging reactions largely involve disassembly of iron-sulphur clusters in proteins. H2O2 or O-2 alone lacked reactivity toward iron regulatory protein-1 (IRP-1), but a combined action of the two species induced reversible inactivation of IRP-1. Such an effect was attributed to direct interactions of O-2 and H2O2 with a preformed pool of IRP-1, resulting in reversible modifications of -SH residues; in fact, its action would be limited to removing only iron atoms, an effect sufficient to abolish enzyme activity.
The hydroxyl radical (.OH) is the most reactive of the free radical molecules. OH- damages cell membranes and lipoproteins by a process termed lipid peroxidation. In fact, lipid peroxidation can be defined as the process whereby free radicals “steal” electrons from the lipids in our cell membranes, resulting in cell damage and increased production of ROS. This process takes place in 3 stages:
Initiation: In a peroxide-free lipid system, the initiation of a peroxidation sequence refers to the attack of an ROS (with sufficient reactivity) able to abstract a hydrogen (H) atom from a methylene group (- CH2-).
Propagation: A peroxyl radical is able to abstract H from another lipid molecule (adjacent fatty acid), especially in the presence of metals such as copper or iron, thus causing an autocatalytic chain reaction. The peroxyl radical combines with H to give a lipid hydroperoxide (or peroxide).
Termination: formation of a hydroperoxide. Lipid peroxidative damage to lipids in low-density lipoprotein (LDL) plays an important role in atherosclerosis [9]. To protect against oxidative damage, organisms have developed a variety of antioxidant defenses that include proteins, compounds such as vitamins, and specialized antioxidant enzymes.
Lipid-soluble antioxidants are located in the cellular membranes and lipoproteins, whereas the water-soluble antioxidants are present in the aqueous environments, such as fluids inside cells and in the blood. Preventative antioxidant enzymes inside the cell are an important defense against free radicals.
In humans, the highest levels of SOD are found in the liver, adrenal gland, kidney, and spleen. Catalase and glutathione peroxidase both work to detoxify O2-reactive radicals by catalyzing the formation of H2O2 derived from O2 -. The liver, kidney, and red blood cells possess high levels of catalase, which helps to detoxify chemicals in the body. The water-soluble tripeptide-thiol glutathione also plays an important role in a variety of detoxification processes. Glutathione is found in millimolar concentrations in the cell cytosol and other aqueous phases, and readily interacts with free radicals, especially the hydroxyl radical, by donating a hydrogen atom.
Sepsis and Signaling Pathways
Serious infections trigger systemic inflammatory response and can result in sepsis. It is believed that sepsis and therefore septic shock are due to the inappropriate increase in the innate immune response via circulating and tissue inflammatory cells, such as monocytes/macrophages and neutrophils. These cells normally exist in a nonactivated state but are rapidly activated in response to bacteria. Sepsis induces a dysfunction in immune cells that contributes to the development of injuries by producing mediators such as cytokines and ROS.
Lipopolysaccharide (Lps) Signaling
LPS of Gram-negative organisms induces macrophages to secrete cytokines, which in turn activate T, and B cells to upregulate the adaptive immune responses. Toll-like receptor 4 (TLR4) is the LPS receptor and its stimulation induces nuclear factor kB (NF-kB) activation. The activation of NF-kB involves phosphorylation and degradation of IkB, an inhibitor of NF-kB. The NF-kB/IkB system exerts transcriptional regulation on proinflammatory genes encoded for various adhesion molecules and cytokines. Activation of NF-kB leads to the induction of NF-kB binding elements in their promoter regions and also leads to the induction of NF-kB dependent effector genes, which produce modifications in blood flow, and aggregation of neutrophils, and platelets. This results in damaged endothelium and also coagulation abnormalities often seen in patients with sepsis and septic shock. Therefore, NF-kB is reported to be an O2 sensor in LPS-induced endotoxemia.
Free Radicals and Antioxidants In Sepsis
The sources of ROS during sepsis are:
the mitochondrial respiratory chain.
the metabolic cascade of arachidonic acid.
the protease-mediated enzyme xanthine oxidase.
granulocytes and other phagocytes activated by complement, bacteria, endotoxin, lysosomal enzymes, etc.
Other oxidases mainly NADPH oxidase.
Under normal physiological conditions, the majority of ROS are formed during cellular respiration and by activated phagocytic cells, including neutrophils, involved in the inflammatory response. ROS have physiologically essential roles in mitochondrial respiration, prostaglandin production pathways and host defense . The electron reduction of O2 occurs in the mitochondrial electron transport system of all aerobically respiring cells. The enzyme catalyzing this transition metals iron and copper in its active site. These ions can be paramagnetic and contain stable unpaired electrons. By using the unpaired electrons in these transition metals to control the O2 reactions, mitochondria prevent the unwanted release of ROS.
In sepsis, there are several potential sources of ROS, including the mitochondrial respiratory electron transport chain, xanthine oxidase activation as a result of ischemia and reperfusion, the respiratory burst associated with immune cell activation, arachidonic acid metabolism and NADPH oxidase.
In fact, activated immune cells produce O2 – as a cytotoxic agent as part of the respiratory burst via the action of membrane-bound NADPH oxidase on O2.
The increase of ROS after LPS challenge has been demonstrated in different models of septic shock in peritoneal macrophages and lymphocytes.
This disturbance in the balance between pro-oxidants (ROS) and antioxidants in favor of the former is characteristic of oxidative stress in immune cells in response to endotoxin. In this context,
a typical behavior of these cells under an oxidative stress situation implies changes in different immune functions such as an increase in adherence and phagocytosis and a decrease in chemotaxis.
Neutrophils play a crucial role in the primary immune defense against infectious agents,which includes phagocytosis and the production of ROS.
Antioxidant Defenses
Antioxidants are central to the redox balance in the human body. They do not act in isolation, but synergistically with other classes of molecules. Primary antioxidants prevent oxygen radical formation, by either removing free radical precursors or by inhibiting catalysis, e.g. the enzymes glutathione peroxidase and catalase. Secondary antioxidants react with ROS which have already been formed, either to remove or inhibit them, e.g. vitamins C and E. Endogenous antioxidant defenses exist in a number of locations, namely intracellularly, on the cell membrane and extracellularly. The immune system is highly reliant on accurate cell-cell communication for optimal function, and any damage to the signaling systems involved will result in an impaired immune responsiveness.
Oxidant-mediated tissue injury is a particular hazard to the immune system, since phagocyte cells produce ROS as part of the defense against infection.
Therefore, adequate amounts of neutralizing antioxidants are required to prevent damage to the immune cells themselves.
The SOD enzymes are a family of metalloenzymes which rapidly promote the conversion of O2- to H2O2. Three forms of SOD are recognized to be important: copper-zinc SOD (cytoplasmic-located), manganese SOD (mitochondrial-located) and extracellular SOD (extracellular matrix-located). Catalase and glutathione peroxidase, a selenium containig enzyme which requires the presence of reduced GSH for its action, both catalyze the conversion of H2O2 to H 2O. GSH also has direct antioxidant activity, through donation of hydrogen ions, to repair damaged DNA. Oxidative stress and modulation on GSH/GSSG (GSSG=oxidized GSH) levels also up-regulate gene expression of several other antioxidant proteins, such as manganese SOD, glutathione peroxidase, thioredoxin (Trx) and metallothionein.
Effects of Nitric Oxide
NO is synthesized from L-arginine by different isoenzymes of (NOS), and is implicated in a wide range of disease processes, exerting both detrimental and beneficial effects at the cellular and vascular levels. To date, three main isoforms of NOS are known:
neuronal NOS (NOS-1 or nNOS),
inducible NOS (NOS-2 or iNOS), and
endothelial NOS (NOS-3 or eNOS).
NO has been shown to play a key role in the pathogenesis of septic shock
Hyperproduction of NO induces
excessive vasodilation,
changes in vascular permeability, and
inhibition of noradrenergic nerve transmission,
all characteristics of human septic shock.
The recogniton of NO production by activated macrophages as part of the inflammatory process was an important milestone for assesing both the biological production of NO and the phenomenon of induction of NOS activity. The observation has been extended to neutrophils, lymphocytes, and other cell types. The role of NO in the pathophysiology of endotoxic shock was advanced by Thiemermann and Vane, who observed that administration of the specific NOS inhibitor N-methyl-L-arginine (L-NMMA) decreased the severe hypotension produced by administration of LPS. Other groups simultaneously reported similar results indicating that endotoxin increases NO production and prompted the idea that pharmacological inhibition of NOS may be useful in the treatment of inflammation and septic shock. However, clinical trials using L-NMMA failed to show a beneficial effect in septic shock patient. The major limitation for the use of NOS inhibitors in clinical studies is the development of pulmonary hypertension as a side effect of NOS blockade, which can be alleviated by the use of inhaled NO.
However, several compounds which modulate NO synthesis have been patented in recent years, such as various inflammatory mediators that have been implicated in the induction and activation of iNOS, particularly IFNg, TNFa, IL-1b, and platelet-activating factor (PAF) alone or synergistically. In addition to the activation of iNOS, cytokines and endotoxin may increase NO release by increasing arginine availability through the opening of the specific y+ channels and the expression of the cationic amino acid transporter (CAT), or by increasing tetrahydrobiopterin levels, a key cofactor in NO synthesis. Several experimental studies have demonstrated a decrease in NOS activity resulting in an impairment in endothelial-dependent relaxation during endotoxemia and experimental sepsis, possibly as the result of a cytokine-or hypoxia-induced shortened half-life of NOS mRNA, or of altered calcium mobilization.
NO exerts in vitro toxic effects including nuclear damage, protein and membrane phospholipid alterations, and the inhibition of mitochondrial respiration in several cell types. Mitochondrial impairment could also be considered as an adaptive phenomenon, decreasing cellular metabolism when the energy supply is limited. The toxicity of NO itself may be enhanced by the formation of ONOO- from the reaction of NO with O-2. Therefore, the multiple organ failure syndrome (MOFS) that often accompanies severe sepsis may be related to the cellular effects of excess NO or ONOO-.
Involvement of Nitrogen Species
NO reacts rapidly with ferrous iron, and at physiological concentrations, NO also binds to soluble guanylate cyclase and to another hemoprotein, cytochrome c oxidase (Complex IV), the terminal enzyme of the mitochondrial respiratory chain. NO can therefore control cellular functions via the reversible inhibition of respiration. There are a number of reactive NO species, such as
N2O3 and
ONOO-
that can also alter critical cellular components.
During the first hours after injury, iNOS-mediated NO production is upregulated, producing a burst of NO that far exceeds basal levels. This overabundance of NO produces significant cellular injury via several mechanisms.
NO may
directly promote overwhelming peripheral vasodilation, resulting in vascular decomposition;
NO may upregulate the transcription NF-kB initiating an inflammatory signaling pathway that, in turn,
triggers numerous inflammatory cytokines.
NO also interacts with the O-2 to yield ONOO-, a highly reactive compound that exacerbates the injury produced by either O-2 alone or NO alone.
The ONOO- generation which occurs during fluid resuscitation in the injured subject produces cellular death by enhancing DNA single strand breakage, activates the nuclear enzyme polyADP ribose synthetase (PARS), leading to cellular energy depletion and cellular necrosis. The detrimental effects of ONOO- in shock and resuscitation have been attributed to oxidation of sulfhydryl groups, the nitration of tyrosine, tryptophane, and guanine, as well as inhibition of the membrane sodium-potassium adenosine triphosphatase. PARS activation depletes NAD and thus alters electron transport, ATP synthesis, and glycolysis; and leads to DNA fragmentation and cellular apoptosis.
The activation of monocytes, macrophages and endothelial cells by LPS results in the expression of iNOS, and consequently increases the transformation of L-arginine to NO, which can combine with O2- to form ONOO-, causing tissue injury during shock, inflammation and ischemia reperfusion. NO stimulates H2O2 and O-2 production by mitochondria, increasing leakage of electrons from the respiratory chain. H2O2, in turn, participates in the upregulation of iNOS expression via NFkB activation. ONOO- has been shown to stimulate H2O2 production by isolated mitochondria. On the other hand, NO can decrease ROS-produced damage that occurs at physiological levels of NO. The high reactivity of NO with radicals might be beneficial in vivo by scavenging peroxyl radicals and inhibiting peroxidation. ONOO- may also be a signal transmitter and can mediate vasorelaxation, similarly to NO.
Local generation of RNS contributes to tissue injury. Recent studies have demonstrated that activation of the nuclear enzyme poly(ADP-ribose) polymerase-1 by RNS-mediated DNA damage is an important pathway of tissue injury in conditions associated with oxidative stress. Increased formation of RNS in response to endotoxin challenge is organ specific.
In sepsis, NO may exert direct and indirect effects on cardiac function. Sustained generation of NO occurs in systemic inflammatory reactions, such as septic shock with involvement in circulatory failure. In fact, myocardial iNOS activity has been reported in response to endotoxin and cytokines and inversely correlated with myocardial performance. Low-to-moderate doses of iNOS inhibitors restore myocardial contractility in hearts exposed to proinflammatory cytokines, whereas at higher doses, the effects are reversed. This finding may indicate that small amounts of NO produced by iNOS may be necessary to maintain contractility and can be cardio-protective in experimental sepsis.
Nitric oxide in Septic Shock
A list of effects of NO in sepsis is as follows.
Inhibition of nitric oxide synthesis causes myocardial ischemia in endotoxemic rats
Nitric oxide causes dysfunction of coronary autoregulation in endotoxemic rats
Prolonged inhibition of nitric oxide synthesis in severe septic shock
Effect of L-NAME, an inhibitor of nitric oxide synthesis, on cardiopulmonary function in human septic shock
Pulmonary hypertension and reduced cardiac output during inhibition of nitric oxide synthesis in human septic shock
Effect of L-NAME, an inhibitor of nitric oxide synthesis, on plasma levels of IL-6, IL-8, TNF-u and nitrite/nitrate in human septic shock
Endothelin-1 and blood pressure after inhibition of nitric oxide synthesis in human septic shock
Distribution and metabolism of NO-nitro-L-arginine methyl ester in patients with septic shock
The possible involvement of the L-arginine-NO pathway in both the vascular and cellular processes seen in sepsis has been supported by numerous in vitro and in vivo studies. iNOS appears to be expressed in a wide array of cell types during sepsis, including immune cells (such as macrophages, neutrophils, T lymphocytes), as well as cells outside the classical immune system (for example, hepatocytes, Kuppfer cells, vascular smooth muscle cells, endothelial cells, and fibroblasts). Expression of iNOS is regulated, both positively and negatively, by a number of mediators present during infection and inflammation. The main stimuli for iNOS induction indude lipopolysaccharide (LPS), interferon-y, interleukin (IL)-10, and tumor necrosis factor (TNF)-a; inhibitory cytokines, such as transforming growth factor-5, IL-4 and IL-10, as well as glucocorticoids, can prevent this induction. The expression of iNOS in response to these agents differs among cell types, but a maximal inducing effect is generally obtained by the combination of microbial products and cytokines acting synergistically.
iNOS activity is also regulated by substrate and cofactor availability. Tetrahydrobiopterin (BH4), an essential cofactor for the enzyme, is coinduced with iNOS in cytokine-stimulated vascular smooth muscle cells.
NO is a simple molecule, but its widespread production in sepsis, coupled with its effects on a variety of intracellular and extracellular target molecules, results in a complex array of biologic roles. Interaction of NO with the metalloproteins in a number of key enzymes can modulate their activity. Many of the signaling actions of ‘NO are mediated by soluble guanylate cyclase. By binding the iron on the heme component of soluble guanylate cyclase, NO is able to activate the enzyme leading to cyclic guanosine monophosphate (cGMP) formation.
Increased cGMP levels account for several of the important cellular actions of NO, including
smooth muscle relaxation,
platelet aggregation and adherence, as well as
neutrophil chemotaxis.
However, *NO can adversely affect cellular metabolism through its disruption of iron-sulfur clusters in essential energy-generating enzymes involved in mitochondrial electron transport, glycolysis, and the Krebs cycle. Further, high concentrations of induced macrophage produced NO can directly interfere with DNA in target cells, resulting in fragmentation.
Another critical reaction that ‘NO undergoes during inflammation is with the superoxide anion radical (02j, yielding peroxynitrite (OONO-). OONO- is a potent oxidant that can decay under acidic conditions to produce a powerful hydoxyl-like free radical. This reaction between *NO and O2 can have a protective or damaging consequence, depending on the individual sites and rates of production of the free radicals, and the redox status of both the generating cells as well as the target cells. OONO- formation can initiate adverse effects such as lipid peroxidation of membranes, and modification of structural proteins through nitration of tyrosine residues (14). Indeed, increased levels of 3-nitrotyrosine have been detected in the lungs of patients with sepsis and animals with acute lung injury. However, OONO- can also S-nitrosylate glutathione and other thiol-containing substances to form S-nitrosothiols, which have marked cardioprotective and cytoprotective effects.
The damaging effect of NOS inhibition may be, in part, mediated by oxygen radicals and platelet deposition, suggesting a cytoprotective role of NO in preventing microvascular thrombosis and as a free radical scavenger. In addition, ‘NO has a protective role in hepatic microcirculatory dysfunction during sepsis through its effect on leukocyte adherence to sinusoidal walls. ‘NO may also protect against circulatory vasoconstrictors during inflammation, as enhanced ‘NO synthesis counteracted phenylephrine-induced increases in intrahepatic resistance in endotoxin-treated rats. Finally, we have recently demonstrated that different types of NOS inhibitors resulted in detectable apoptosis in the liver following LPS injection. This increase in apoptosis was present even with L-N-iminoethyl-lysine (L-NIL), a rather specific inhibitor of iNOS, revealing another important protective role of NO as an antiapoptotic agent in sepsis.
Even though overproduction of *NO in the vasculature contributes to the vasodilatation seen in septic shock, iNOS expression during inflammation also represents a beneficial, adaptive response in some organ systems. Moreover, different tissues can react dissimilarly to the effects of ‘NO cytotoxicity. In this setting, global nonselective inhibition of NOS, including the potentially undesirable consequences of eNOS inhibition, would be harmful. If confirmed, this would suggest that use of isoform-specific inhibitors of NOS within the vascular bed would be more appropriate.
Pulmonary Hypertension and Reduced Cardiac Output
Pulmonary hypertension and reduced cardiac output can be major side effects of continuous NO synthase inhibition. Pulmonary vasoconstriction is undesirable because it may compromise pulmonary gas exchange and because it increases the workload on the right ventricle. In cases where strain already exists on the right ventricle (e.g. sepsis or PEEP ventilation) or in cases where right sided cardiac reserve is minimal, such increase in workload may lead to right ventricular failure, reduced cardiac output and compromised tissue perfusion.
Blood pressure and systemic vascular resistance increased during infusion of the NO synthase inhibitor L-NAME, and the dosage of catecholamines was reduced. The vasoconstrictive response to L-NAME most likely was the result of blocking the NO system . In addition to the systemic effects of L-NAME, severe pulmonary vasoconstriction was observed with L-NAME. Analogous to these findings, in patients with Adult Respiratory Distress Syndrome (ARDS), inhalation of NO is reported to be beneficial by causing local vasodilation in bronchial and pulmonary circulation which results in reduced pulmonary vascular resistance and improved oxygenation. This suggests that the pulmonary circulation is sensitive to the vasodilating effects of both endogenous and exogenous NO. Pulmonary vasoconstriction is not, therefore, unexpected with systemic inhibition of NO synthesis. With a continuous infusion of L-NAME, pulmonary vascular resistance increased five-fold, whereas systemic vascular resistance “only” doubled. pulmonary hypertension was reversible after stopping L-NAME infusion. In prior experiments with a lower dose of LNAME, pulmonary vasoconstriction was less pronounced and did not result in pulmonary hypertension.’ Thus, pulmonary hypertension is a dose-related effect of L-NAME that can probably be attributed to overdosing of the drug. Reduced cardiac output may have directly resulted from the extreme increase in pulmonary vascular resistance compromising venous return and left ventricular preload and/or a reflex reduction in heart rate by the increase in vascular resistance and blood pressure. S-methyl-isothiourea, a relatively selective inhibitor of iNOS activity, decreased pulmonary leak and improved survival in endotoxemia. However, because of the tissue-protective and antiapoptotic effects of NO, even selective iNOS inhibitors may be detrimental in certain tissues during sepsis.Combining the salutary effects of site-specific local donors that exploit the cytoprotective actions of ‘NO with specific agents that combat the deleterious hypotensive and tissue-damaging effects of ‘NO overproduction may be needed to treat septic shock. In this regard, inhaled ‘NO gas has shown promise as a selective pulmonary vasodilator.
Role of Nitric Oxide in Inflammation and Tissue Injury
Since the discovery that nitric oxide (‘NO) accounts for the biologic activity of endothelial-derived relaxing factor, a torrent of research over the last decade has focused on its role, protective or detrimental, in myriad pathophysiologic conditions. Recently, increasing attention has focused on ‘NO as a possible mediator of the severe hypotension and impaired vasoreactivity characteristic of circulatory failure. Experimental and clinical studies have suggested NOS inhibition might have therapeutic potential in circulatory shock, and other studies have demonstrated the beneficial nature of iNOS expression in modulating tissue perfusion and mediating cytotoxicity. However, inhibition of ‘NO synthesis in experimental and clinical studies of shock has yielded mixed, sometimes contradictory, results. Overproduction of ‘NO in the vasculature may result in systemic vasodilatation, but still ‘NO synthesis has a beneficial role in regulating organ perfusion and mediating cytotoxicity.
Diminished *NO Production Occurs with Hemorrhage
These findings are consistent with those in trauma patients, where nitrite and nitrate levels were reduced for prolonged periods after injury. This impairment of ‘NO production in victims of hemorrhagic hypotension may be due to impairment of eNOS, and indeed, several investigators have demonstrated decreased vasodilatory activity in vascular rings taken from hemorrhaged animals in response to agonists that stimulate endothelial ‘NO production. In studies of hemorrhagic shock no iNOS expression could be detected until the very late irreversible phase of HS. The hemodynamic instability associated with decompensation occurred well before NOS induction.
Using either the selective inhibitor L-NIL or iNOS knockout mice, iNOS inhibition or deficiency not only prevented the upregulation of the inflammatory cytokines IL-6 and granulocyte colony-stimulating factor following resuscitation from HS but also produced a marked reduction in lung and liver injury. Furthermore, the activation of the proinflammatory transcriptional factors nuclear factor kappa B and signal transducer and activator of transcription 3 was also reduced, suggesting iNOS upregulation has a key role in proinflammatory signaling and the subsequent activation of inflammatory cascades. Recent studies have implicated a possible redox-sensitive mechanism. ‘NO activates the critical signaling enzyme p21ras through S-nitrosylation.
Vascular quenching of ‘NO using scavengers may again provide an alternative to NOS inhibition as a means to achieve the goal of reducing ‘NO levels. Use of ‘NO scavengers after HS and resuscitation may serve to supplement a possibly depleted antioxidant defense system and limit the harmful effects of free radicals such as OONO- and hydoxyl radicals. Removal of ‘NO by this method is complicated by the extreme rapidity of the reaction between ‘NO and 02’- .
iNOS upregulation also has a beneficial protective role in several organ systems. In conditions where excess NO production results in maladaptive damaging consequences with disruption of homeostasis, the therapeutic strategy should be to remove this surplus ‘NO without adversely affecting the cytoprotective actions of *NO. Interfering with the physiologic and microcirculatory role of eNOS through nonselective, global inhibition of NOS is undesirable in shock.
Effects of nitric oxide in endotoxemia and hemorrhagic shock and proposed therapeutic strategies for manipulation of nitric oxide production.
Endotoxemia Hemorrhagic shock
Effects of NO Beneficial Beneficial
by eNOS -maintains perfusion; -maintains perfusion;
cytoprotective cytoprotective
iNOS Beneficial and toxic- Beneficial and toxic-
depending on site can induce tissue damage and of production and promote inflammation with microenvironment sustained shock
Therapeutic strategy
Inhibition of eNOS Avoid Avoid
Inhibition of iNOS Possibly desirable- Probably desirable-
to reduce to limit exaggerated inflammatory
cytotoxicity and response and development of
combat hypotension MODS
NO scavengers Probably desirable-quench Probably desirable-quench
extracellular NO without extracellular NO without
inhibition of eNOS or iNOS; inhibition of eNOS or iNOS;
supplement antioxidant defenses supplement antioxidant defenses
NO donors Possibly desirable-site- Possibly desirable-site-
specific donors without adverse specific donors without adverse
systemic side effects; systemic side effects;
limited availability limited availability
Therapeutic Outlook
LINCS: L-NAME (a NO synthase inhibitor)
Patients were randomized to supportive care alone (n=15, control group) or to supportive care in addition to L-NAME (1 mg/Kg bolus and 1 mg/Kg/h continuous IV drip for 5 h n=15). Death at one month was 27% in the L-NAME group vs. 67% in the control group (p=0.008). Time on IABP and time on mechanical ventilation were significantly shorter in the L-NAME group. The results of this study indicate that NO synthase inhibitors are beneficial in the treatment of patients with refractory cardiogenic shock. Inducible Nitric Oxide Synthase Inhibitors
Inducible nitric oxide synthase (iNOS)-dependent production of nitric oxide (NO) plays an important role in inflammation. The effects of various naturally occurring furanocoumarins on NO production in lipopolysaccharide (LPS)-activated RAW 264.7 macrophage cells were evaluated in vitro. The results showed that angelicin, pimpinellin, sphondin, byakangelicol, oxypeucedanin, oxypeucedanin hydrate, xanthotoxin, and cnidilin are potential NO production inhibitors, and their IC50 values for inhibition of nitrite production were 19.5, 15.6, 9.8, 16.9, 16.8, 15.8, 16.6, and 17.7 mg/mL, respectively.
Distinct structure activity relationships were also revealed for the NO production inhibitory activities of these furanocoumarins. Activities of the angelicin type such as pimpinellin and sphondin were more potent than those of the psoralen type. Presence of a methoxy at the C6 position in the angelicin type seemed to be essential to augment the activity. Western blot analysis demonstrated that only sphondin dose-dependently inhibited the expression of the iNOS protein at 2.5±20 mg/mL. However, iNOS enzyme activity was stimulated with LPS for 12 h and sphondin was administered (20 mg/mL) for 24 h, which did not reasonably inhibit iNOS enzyme activity. l-NAME (100 mM), a known specific inhibitor of iNOS, was employed as a positive control with the same protocol and showed more than 50% inhibition activity. The results demonstrate that the NO production inhibitory activity of sphondin is due to the effect of iNOS expression, but not by direct inhibition of iNOS enzyme activity. Thus, sphondin may act as a potent inhibitor of NO production under tissue-damaging inflammatory conditions. S-Methylisothiourea Sulfate, A Potent And Selective Inhibitor Of Inducible Nitric Oxide Synthase
Non-isoform-selective inhibition of NO formation, however, may lead to side effects by inhibiting the constitutive isoform of NOS and, thus, the various physiological actions of NO. S-Methylisothiourea sulfate (SMT) is at least 10- to 30-fold more potent as an inhibitor of inducible NOS (iNOS) in immuno-stimulated cultured macrophages (EC50, 6 ,AM) and vascular smooth muscle cells (EC50, 2 ,uM) than NG-methyl-L-arginine (MeArg) or any other NOS inhibitor yet known. The effect of SMT on iNOS activity can be reversed by excess L-arginine in a concentration-dependent manner.
Enhanced formation of NO following the induction of iNOS contributes importantly to the circulatory failure (hypotension and vascular hyporeactivity to vasoconstrictor agents) in circulatory shock of various etiologies.
SMT dose-dependently reverses (0.01-3 mg/kg) the hypotension and the vascular hyporeactivity to vasoconstrictor agents caused by endotoxin [bacterial lipopolysaccharide]
SMT, a potent and selective inhibitor of iNOS, may have considerable value in the therapy of circulatory shock of various etiologies and other pathophysiological conditions associated with induction of iNOS. SMT, or other iNOS-selective inhibitors, are likely to have fewer side effects which are related to the inhibition of eNOS, such as excessive vasoconstriction and organ ischemia), increased platelet and neutrophil adhesion and accumulation, and microvascular leakage.
Iron Chelates Bind Nitric Oxide
Nitric oxide (NO), a short-lived potent vasodilator, was first described as the endothelium-derived relaxation factor (EDRF). The formation of NO from the guanidine nitrogen group of L-arginine is catalyzed by a group of enzymes termed constitutive (cNOs) and inducible (iNOs) NO synthases. The inducible form is not present constitutively in mammalian cells but is induced by proinflammatory stimuli such as bacterial lipopolysaccharide (LPS), Corynebacterium parvum, and the cytokines tumor necrosis factor-a, interleukin-1, or interferon-y, individually or in combination. Excess production of NO is reported to be associated with the development of hypotension associated with endotoxemia and sepsis.
Electrochemical studies show that FeIII-(DTPA)2- binds NO stoichiometrically upon reduction to iron(II) at biologically relevant potentials to form a stable NO adduct. In contrast, FeI”I(HDFB)+ is a stable and efficient electrocatalyst for the reduction of NO to N20 at biologically relevant potentials. These results suggest that the mechanism of protection against death by septic shock involves NO scavenging and that particularly effective drugs that operate a low dosages may be designed based on the principle of redox catalysis. These complexes constitute a new family of drugs that rely on the special ability of transition metals to activate small molecules.
Iron complexes could act as general NO scavengers and provide protection against septic shock. Iron complexes are capable of forming relatively stable NO adducts. Metal complexes, and in particular iron chelators, could act as “molecular sponges,” mopping up the excess NO produced during septic shock. Iron chelators can sequester and (as for 2) catalyze conversion of NO to benign products. Demonstration of mechanistic aspects of septic shock protection in vivo, including interaction with other free radicals, may be hampered by the detection limits of current analytical techniques. To detect the NO Fe-DETC complex formation in livers of LPS-treated mice by the electron paramagnetic resonance.
After screening a library of metal chelators and chelates [Fe(III)(H2DTPA)] and [Fe(III)(HDFB)]+ offered the highest mortality decrease in an experimental model of septic shock. The Fe(II) form of both complexes can bind NO, which appears to be related to their biological function.
Survival was greatly enhanced by the administration of 4 or 2 either 2 h before and at the time of or 30 min after LPS. In contrast, the Fe3+-free ligands of these compounds, 3 and 1, were less protective when administered before and at the time of LPS and virtually ineffective when administered after LPS. The clear advantage of 4 over 2 when administered after LPS was observed over a large number of experiments [76% survival with 4 (n = 102 mice) and 38% survival with 2 (n =64 mice)].
The hydroxamic acid siderophore ferrioxamine B [Fe”‘(HDFB)+] and the iron complex of diethylene-triamine-pentaacetic acid [FeI”(DTPA)2i] protected mice against death by septic shock induced by Corynebacterium parvum + lipopolysaccharide. Although Fem(DTPA)2- was somewhat more effective than FeI”(HDFB)+, the iron-free ligand H4DFB+ was significantly more effective than DTPA. The hydroxamic acid chelator has a much higher iron affinity than the amine carboxylate, allowing for more efficient formation of the FeI”(HDFB)+ complex upon administration of the iron-free ligand.
Efficacy of Treatment With the Iron (III) Complex of Diethylenetriamine Pentaacetic Acid
Bacteremia and septic shock also are associated with overproduction of free radicals such as hydroxyl, superoxide, and carbon- and oxygen-centered radicals. In addition, nitric oxide (NO) overproduction is at least partly responsible for the vasodilation that causes a reduction in mean systemic arterial pressure (MSAP) and organ perfusion pressure during septic shock. This overproduction of NO likely results from early activation of the endothelial constitutive form of NO synthase followed by induction of the inducible form of NO synthase via TNF and IL-1.
The simultaneous increase and further reaction of NO with superoxide, which yields the oxidant peroxynitrite anion, occurs in cellular systems in response to inflammatory mediators. In addition, in sepsis-associated adult respiratory distress syndrome (ARDS), the presence of nitrotyrosine residues (formed by reaction of peroxynitrite and the tyrosine residues of proteins) are apparent throughout the lung.
Administration of the iron (III) complex of diethylenetriamine pentaacetic acid (DTPA iron (III), prevented death in Corynebacterium parvum 1 LPS-treated mice. Using electrochemistry, the binding of NO to DTPA iron (II) is confirmed. The DTPA iron (II) form can be easily formed by common biological reductants, because the potential for the iron (III/II) couple is E = 0.22.
Treatment with DTPA iron (III) resulted in a significant decrease in mortality compared to the untreated controls. The efficacy of DTPA iron (III) increased when given to mice 2 h or more after infection. The best results were observed when DTPA iron (III) was given 5 h after infection.
The iron (III) complex of diethylenetriamine pentaacetic acid (DTPA iron [III]) protected mice and baboons from the lethal effects of an infusion with live LD 100 Escherichia coli. In mice, optimal results were obtained when DTPA iron (III) was administered two or more hours after infection. Prevention of death occurred in spite of the fact that the adverse effects of TNFa were well underway in the mouse model.
In septic baboons, survival was observed after administration of two doses of DTPA iron (III) at 2.125 mg/kg, the first one given before, or as late as 2 h after, severe hypotension. Administration of DTPA iron (III) did not alter mean systemic arterial pressure, but did protect baboons in the presence of high levels of TNFa and free radical overproduction. Furthermore, exaggerated production of nitric oxide was attenuated. Because of its ability to interact in vitro with free radicals, its poor cell permeability, and its short half-life, we postulate that DTPA iron (III) and/or its reduced form may have protected the mice and baboons by sequestration and subsequent elimination of free radicals (including nitric oxide) from their systems. (J. Clin. Invest.1996. 98:192–198.) Inhibitor of Poly(Adenosine 5′-Diphosphate-Ribose) Synthetase
Poly(adenosine 5′-diphosphate [ADP]-ribose) synthetase (PARS) is a nuclear enzyme which, when activated by DNA singlestrand breaks, initiates an energy-consuming, inefficient metabolic cycle by transferring ADP-ribose units to nuclear proteins. The result of this process is a rapid depletion of intracellular oxidized nicotinamide adenine dinucleotide and adenosine 5′-triphosphate energetic pools, which slows the rate of glycolysis and mitochondrial respiration, leading to cellular dysfunction and death.
Reactive oxygen-centered radicals (superoxide, hydroxyl radicals, singlet oxygen, and hydrogen peroxide) and peroxynitrite (a reactive oxidant produced from the reaction of superoxide and nitric oxide) are powerful triggers of DNA single strand breakage, and they induce activation of a cell suicide cycle governed by PARS in various cell types in vitro.
Multiple reports implicated a role of PARS activation in the pathophysiology of endotoxic shock, hemorrhagic shock, and various forms of ischemia-reperfusion injury.
Twenty pigs were chronically instrumented with intracardiac transducers to measure left ventricular pressure, sonomicrometer crystals in the left ventricle to measure short axis diameter, an ultrasonic flow meter to measure cardiac output, and catheters in the pulmonary artery and aorta to measure blood pressures and collect samples. By using a randomized study design, either the novel potent PARS inhibitor PJ34 (10 mg/kg for 1 hr, 2 mg·kg 1·hr 1 for 96 hrs) or placebo to pigs immediately before intraperitoneal implantation of Escherichia coli 0111.B4 (2.3 0.1 1010 colony-forming units/kg)-laden fibrin clots to produce peritonitis and bacteremia.
PJ34 treatment significantly attenuated this cytokine response. The formation of peroxynitrite and the activation of PARS were confirmed in hearts and lungs of the septic pigs by the immunohistochemical detection of nitrotyrosine and poly(ADP-ribose), respectively. Inhibition of PARS with PJ34 abolished poly(ADP-ribose) formation in septic animals.
Cardiac inotropicity was evaluated by analysis of percentage of short axis diameter shortening (one-dimensional ejection fraction). Bacteremia induced a rapid and progressive loss of inotropy until death in vehicle-treated pigs. A similar decline was observed in the first 6 hrs in PJ34 pigs. This decline was reversed on all subsequent days. Control pigs exhibited rapid and significant increases in systemic vascular (SVR) and pulmonary vascular (PVR) resistances.
This experimental model mimics many aspects of the human sepsis syndrome. Therefore, the positive survival benefit of PARS inhibition suggests a potential utility of PARS inhibitors in human sepsis management. PARS activation is triggered by DNA single-strand breakage.
The current work, demonstrating increased poly(ADP-ribose) staining in the heart of septic pigs, may point toward the importance of a myocardial, PARS dependent cardiodepressive mechanism in the current model of shock. This hypothesis is supported by the following findings:
free radicals cause myocardial dysfunction and injury in a PARS dependent fashion in vitro;
in the current study, pharmacologic inhibition of PARS with PJ34 markedly improved myocardial function; and
in prior studies, pharmacologic inhibition of PARS markedly improved myocardial contractile function in hypoxic-reoxygenated hearts as well as in a porcine model of hemorrhagic shock.
Treatment with a potent PARS inhibitor improved survival and cardiovascular status and attenuated an important mediator component of the inflammatory response in a lethal porcine model of sepsis. (Crit Care Med 2002; 30:974 –980).
Decrease of the inflammatory response and induction of the Akt/protein kinase B pathway by poly-(ADP-ribose) polymerase 1 inhibitor
The lack of efficacy of anti-inflammatory drugs, anti-coagulants, anti-oxidants, etc. in critically ill patients has shifted interest towards developing alternative treatments. Since inhibitors of the nuclear enzyme poly-(ADP-ribose) polymerase (PARP) were found to be beneficial in many pathophysiological conditions associated with oxidative stress and PARP-1 knock-out mice proved to be resistant to bacterial lipopolysaccharide (LPS)-induced septic shock, PARP. The mechanism of the protective effect of a potent PARP-1 inhibitor, PJ34 was studied in LPS-induced (20 mg/kg, i.p.) septic shock in mice.
We demonstrated a significant inflammatory response by magnetic resonance imaging in the dorsal subcutaneous region, in the abdominal regions around the kidneys and in the inter-intestinal cavities. We have found necrotic and apoptotic histological changes as well as obstructed blood vessels in the liver and small intestine. Additionally, we have detected elevated tumor necrosis factor-a levels in the serum and nuclear factor kappa B activation in liver of LPS-treated mice.
Pre-treating the animals with PJ34 (10 mg/kg, i.p.), before the LPS challenge, besides rescuing the animals from LPS-induced death, attenuated all these changes presumably by activating the phosphatidylinositol 3-kinase–Akt/protein kinase B cytoprotective pathway.
PJ34, a novel, potent PARP-1 inhibitor was found to protect against LPS induced tissue damage. PARP inhibitors protected Langendorff-perfused hearts against ischemia-reperfusion induced damages by activating the PI3-kinase–Akt pathway. The importance of the PI3-kinase–Akt pathway in LPS induced inflammatory mechanisms has gained support, raising the question whether this pathway was involved in the effect of PJ34 on LPS-induced septic shock.
Among all the observed LPS-induced inflammatory responses, we found the most characteristic and most pronounced increases in the gastro-intestinal tract, but no signal increase could be observed inside the kidneys and in skeletal muscle, in the paravertebral or in the femoral muscle. All increases in signal intensities were significantly attenuated in mice treated with PJ34.
Effect of PJ34 on survival of LPS-treated mice
PARP-1 inhibitor significantly protected the animals against LPS-induced death, with 86 and 43% surviving mice, respectively.
PJ34 treatment itself did not induce death or any obvious damage.
Effect of PJ34 on LPS-induced NF-kB activation
LPS treatment in the lung caused a significant increase in NF-kB activation that was slightly but not statistically significantly attenuated by PJ34 pre-treatment.
in contrast to the lung, NF-kB activation in the liver was prevented by PJ34 pre-treatment
The other tissue with observable LPS-induced pathological changes was the small intestine. Atrophy of villi may reflect the diarrhea observed in the LPS-treated animals and is in agreement with the results of Abreu et al. who found a Fas-mediated apoptosis in intestinal epithelial cells that was sensitised by inhibitors of PI3-kinase and opposed by expressing constitutively active Akt.
Pre-treatment of the animals with a novel, potent PARP-1 inhibitor, PJ34, diminished the thoracic and abdominal inflammatory responses as revealed by T2 imaging, and abolished the above mentioned pathological changes.
The protective role of PARP inhibitors in septic shock is likely to be more complex than merely the regulation of NF-kB/Rel-dependent gene expression.
Activation of the PI3-kinase–Akt/protein kinase B cytoprotective pathway is likely to contribute to the protective effects of PARP inhibitors in shock and inflammation.
Carboxy-PTIO On Hemodynamic And Blood Gas Changes
Infusion of LPS caused a marked decrease in mean arterial pressure (MAP), metabolic acidosis, and hypoxia. These effects were reversed by co-administration of carboxy-PTIO, without affecting other hemodynamic parameters. In control animals, neither hemodynamic nor blood gas parameters changed with or without carboxy-PTIO.
These results indicate that carboxy-PTIO attenuates
LPS-induced hypotension,
metabolic acidosis, and
hypoxia
by scavenging excess NO from the circulation without affecting NO synthase (NOS) activity. An NO scavenger, carboxy-PTIO, may be preferable to non-selective NOS inhibitors for the treatment of human septic shock.
Asymmetrical Dimethyl Arginine Levels
Overwhelming infection with resultant multiple organ failure, which has been termed the ‘sepsis syndrome’ , is a devastating illness with an incidence of 3 per 1,000 population per annum. It has been characterised as a dysregulation of inflammation in response to infection attributable to a combination of
excessive inflammation,
disseminated coagulopathy and
disruption of the integrity of microvascular endothelium.
Asymmetrical dimethyl arginine (ADMA) is an endogenous non-selective inhibitor of nitric oxide synthase that may influence the severity of organ failure and the occurrence of shock secondary to an infectious insult. Levels may be genetically determined by a promoter polymorphism in a regulatory gene encoding dimethylarginine dimethylaminohydrolase II (DDAH II).
A prospective observational study was designed, and 47 intensive care unit (ICU) patients with severe sepsis and 10 healthy controls were enrolled. Serum ADMA and IL-6 were assayed on admission to the ICU and seven days later. Allelic variation for a polymorphism at position -449 in the DDAH II gene was assessed in each patient.
ADMA levels and Sequential Organ Failure Assessment scores were directly associated on day one (p = 0.0001) and day seven (p = 0.002). The degree of acidaemia and lactaemia was directly correlated with ADMA levels at both time points (p < 0.01). On day seven, IL-6 was directly correlated with ADMA levels (p = 0.006). The variant allele with G at position -449 in the DDAH II gene was associated with increased ADMA concentrations at both time points (p < 0.05). Severity of organ failure, inflammation and presence of early shock in severe sepsis are associated with increased ADMA levels. ADMA concentrations may be influenced by a polymorphism in the DDAH II gene.
Several studies have added to the confusion surrounding the role of NO by demonstrating no effect of NO or NOS inhibition on the myocardium or on b-adrenergic responsiveness. Nevertheless, in most studies,
low-to-moderate doses of iNOS inhibitors restore myocardial contractility in hearts exposed to proinflammatory cytokines, whereas
at higher doses, the effects are reversed.
This finding may indicate that small amounts of NO produced by iNOS may be necessary to maintain contractility and can be cardio-protective in experimental sepsis.
Major cellular sources of ROS in living cells. Novo and Parola Fibrogenesis & Tissue Repair 2008 1:5 doi:10.1186/1755-1536-1-5 (Photo credit: Wikipedia)
endogenously produced VIP and PACAP are participants of the natural anti-inflammatory machinery.
VIP and PACAP are two attractive candidates for the development of therapies against acute and chronic inflammatory diseases, septic shock, and autoimmune diseases
Nitric oxide (NO) is a lipophilic, highly diffusible and short-lived molecule that acts as a physiological messenger and has been known to regulate a variety of important physiological responses including vasodilation, respiration, cell migration, immune response and apoptosis. Jordi Muntané et al
NO is synthesized by the Nitric Oxide synthase (NOS) enzyme and the enzyme is encoded in three different forms in mammals: neuronal NOS (nNOS or NOS-1), inducible NOS (iNOS or NOS-2), and endothelial NOS (eNOS or NOS-3). The three isoforms, although similar in structure and catalytic function, differ in the way their activity and synthesis in controlled inside a cell. NOS-2, for example is induced in response to inflammatory stimuli, while NOS-1 and NOS-3 are constitutively expressed.
Regulation by Nitric oxide
NO is a versatile signaling molecule and the net effect of NO on gene regulation is variable and ranges from activation to inhibition of transcription.
The intracellular localization is relevant for the activity of NOS. Infact, NOSs are subject to specific targeting to subcellular compartments (plasma membrane, Golgi, cytosol, nucleus and mitochondria) and that this trafficking is crucial for NO production and specific post-translational modifications of target proteins.
Role of Nitric oxide in Cancer
One in four cases of cancer worldwide are a result of chronic inflammation. An inflammatory response causes high levels of activated macrophages. Macrophage activation, in turn, leads to the induction of iNOS gene that results in the generation of large amount of NO. The expression of iNOS induced by inflammatory stimuli coupled with the constitutive expression of nNOS and eNOS may contribute to increased cancer risk. NO can have varied roles in the tumor environment influencing DNA repair, cell cycle, and apoptosis. It can result in antagonistic actions including DNA damage and protection from cytotoxicity, inhibiting and stimulation cell proliferation, and being both anti-apoptotic and pro-apoptotic. Genotoxicity due to high levels of NO could be through direct modification of DNA (nitrosative deamination of nucleic acid bases, transition and/or transversion of nucleic acids, alkylation and DNA strand breakage) and inhibition of DNA repair enzymes (such as alkyltransferase and DNA ligase) through direct or indirect mechanisms. The Multiple actions of NO are probably the result of its chemical (post-translational modifications) and biological heterogeneity (cellular production, consumption and responses). Post-translational modifications of proteins by nitration, nitrosation, phosphorylation, acetylation or polyADP-ribosylation could lead to an increase in the cancer risk. This process can drive carcinogenesis by altering targets and pathways that are crucial for cancer progression much faster than would otherwise occur in healthy tissue.
NO can have several effects even within the tumor microenvironment where it could originate from several cell types including cancer cells, host cells, tumor endothelial cells. Tumor-derived NO could have several functional roles. It can affect cancer progression by augmenting cancer cell proliferation and invasiveness. Infact, it has been proposed that NO promotes tumor growth by regulating blood flow and maintaining the vasodilated tumor microenvironment.NO can stimulate angiogenesis and can also promote metastasis by increasing vascular permeability and upregulating matrix metalloproteinases (MMPs). MMPs have been associated with several functions including cell proliferation, migration, adhesion, differentiation, angiogenesis and so on. Recently, it was reported that metastatic tumor-released NO might impair the immune system, which enables them to escape the immunosurveillance mechanism of cells. Molecular regulation of tumour angiogenesis by nitric oxide.
S-nitrosylation and Cancer
The most prominent and recognized NO reaction with thiols groups of cysteineresidues is called S-nitrosylation or S-nitrosation, which leads to the formation of more stable nitrosothiols. High concentrations of intracellular NO can result in high concentrations of S-nitrosylated proteins and dysregulated S-nitrosylation has been implicated in cancer. Oxidative and nitrosative stress is sensed and closely associated with transcriptional regulation of multiple target genes.
Following are a few proteins that are modified via NO and modification of these proteins, in turn, has been known to play direct or indirect roles in cancer.
NO mediated aberrant proteins in Cancer
Bcl2
Bcl-2 is an important anti-apoptotic protein. It works by inhibiting mitochondrial Cytochrome C that is released in response to apoptotic stimuli. In a variety of tumors, Bcl-2 has been shown to be upregulated, and it has additionally been implicated with cancer chemo-resistance through dysregulation of apoptosis. NO exposure causes S-nitrosylation at the two cysteine residues – Cys158 and Cys229 that prevents ubiquitin-proteasomal pathway mediated degradation of the protein. Once prevented from degradation, the protein attenuates its anti-apoptotic effects in cancer progression. The S-nitrosylation based modification of Bcl-2 has been observed to be relevant in drug treatment studies (for eg. Cisplatin). Thus, the impairment of S-nitrosylated Bcl-2 proteins might serve as an effective therapeutic target to decrease cancer-drug resistance.
p53
p53 has been well documented as a tumor suppressor protein and acts as a major player in response to DNA damage and other genomic alterations within the cell. The activation of p53 can lead to cell cycle arrest and DNA repair, however, in case of irrepairable DNA damage, p53 can lead to apoptosis. Nuclear p53 accumulation has been related to NO-mediated anti-tumoral properties. High concentration of NO has been found to cause conformational changes in p53 resulting in biological dysfunction.. In RAW264.7, a murine macrophage cell line, NO donors induce p53 accumulation and apoptosis through JNK-1/2.
HIF-1a
Hypoxia-inducible factor 1 (HIF1) is a heterodimeric transcription factor that is predominantly active under hypoxic conditions because the HIF-1a subunit is rapidly degraded in normoxic conditions by proteasomal degradation. It regulates the transciption of several genes including those involved in angiogenesis, cell cycle, cell metabolism, and apoptosis. Hypoxic conditions within the tumor can lead to overexpression of HIF-1a. Similar to hypoxia-mediated stress, nitrosative stress can stabilize HIF-1a. NO derivatives have also been shown to participate in hypoxia signaling. Resistance to radiotherapy has been traced back to NO-mediated HIF-1a in solid tumors in some cases.
PTEN
Phosphatase and tensin homolog deleted on chromosome ten (PTEN), is again a tumor suppressor protein. It is a phosphatase and has been implicated in many human cancers. PTEN is a crucial negative regulator of PI3K/Akt signaling pathway. Over-activation of PI3K/Akt mediated signaling pathway is known to play a major role in tumorigenesis and angiogenesis. S-nitrosylation of PTEN, that could be a result of NO stress, inhibits PTEN. Inhibition of PTEN phosphatase activity, in turn, leads to promotion of angiogenesis.
C-Src
C-src belongs to the Src family of protein tyrosine kinases and has been implicated in the promotion of cancer cell invasion and metastasis. It was demonstrated that S-nitrosylation of c-Src at cysteine 498 enhanced its kinase activity, thus, resulting in the enhancement of cancer cell invasion and metastasis.
Jaiswal M, et al. Nitric oxide in gastrointestinal epithelial cell carcinogenesis: linking inflammation to oncogenesis. Am J Physiol Gastrointest Liver Physiol. 2001 Sep;281(3):G626-34. http://www.ncbi.nlm.nih.gov/pubmed/11518674
What is Septic Shock?
Scripps Research Professor Wolfram Ruf and colleagues have identified a key connection between the signaling pathways and the immune system spiraling out of control involving the coagulation system and vascular endothelium that, if disrupted may be a target for sepsis. (Science Daily, Feb 29, 2008). It may be caused by a bacterial infection that enters the bloodstream, but we now recognize the same cascade not triggered by bacterial invasion. These invading bacteria produce endotoxins and other toxins that trigger a widespread inflammatory response of the innate immune system–a response that is necessary, as it turns out, because without the inflammation, the body cannot fight off the bacterial infection. During sepsis, the inflammation triggers widespread coagulation in the bloodstream. This coagulation can block blood vessels in vital organs, starving the organs of oxygen and damaging them. The organs can be further damaged when the blood starts to flow again because the lining of the blood vessels remain leaky due to inflammatory cytokines and damage by intravascular coagulation.
What is the Pathogenesis of Sepsis?
The acute respiratory distress syndrome (ARDS) has been defined as a severe form of acute lung injury featuring pulmonary inflammation and increased capillary leak. ARDS is associated with a high mortality rate and accounts for 100,000 deaths annually in the United States. ARDS may arise in a number of clinical situations, especially in patients with sepsis. A well-described pathophysiological model of ARDS is one form of the acute lung inflammation mediated by neutrophils, cytokines, and oxidant stress. Neutrophils are major effect cells at the frontier of innate immune responses, and they play a critical role in host defense against invading microorganisms. The tissue injury appears to be related to proteases and toxic reactive oxygen radicals released from activated neutrophils. In addition, neutrophils can produce cytokines and chemokines that enhance the acute inflammatory response. Neutrophil accumulation in the lung plays a pivotal role in the pathogenesis of acute lung injury during sepsis. Directed movement of neutrophils is mediated by a group of chemoattractants, especially CXC chemokines. Local lung production of CXC chemokines is intensified during experimental sepsis induced by cecal ligation and puncture (CLP). Under these conditions of stimulation, activation of MAPKs (p38, p42/p44) occurs in sham neutrophils but not in CLP neutrophils, while under the same conditions phosphorylation of p38 and p42/p44 occurs in both sham and CLP alveolar macrophages. These data indicate that, under septic conditions, there is impaired signaling in neutrophils and enhanced signaling in alveolar macrophages, resulting in CXC chemokine production, and C5a appears to play a pivotal role in this process. As a result, CXC chemokines increase in lung, setting the stage for neutrophil accumulation in lung during sepsis.
Uncontrolled activation of the coagulation cascade following lung injury contributes to the development of lung inflammation and fibrosis in acute lung injury/acute respiratory distress syndrome (ALI/ARDS) and fibrotic lung disease. This article reviews our current understanding of the mechanisms leading to the activation of the coagulation cascade in response to lung injury and the evidence that excessive procoagulant activity is of pathophysiological significance in these disease settings. This is consistent with a pneumonia or lung injury preceding sepsis. On the other hand, it is not surprising that abdominal, cardiac bypass, and post cardiac revascularization may also lead to events resembling sepsis and/or cardiovascular collapse. The tissue factor-dependent extrinsic pathway is the predominant mechanism by which the coagulation cascade is locally activated in the lungs of patients with ALI/ARDS and pulmonary fibrosis. The cellular effects mediated via activation of proteinase-activated receptors (PARs) may be of particular importance in influencing inflammatory and fibroproliferative responses in experimental models involving direct injury to the lung. In this regard, studies in PAR1 knockout mice have shown that this receptor plays a major role in orchestrating the interplay between coagulation, inflammation and lung fibrosis.
The activation of the coagulation cascade is one of the earliest events initiated following tissue injury. The prime function of this complex and highly regulated proteolytic system is to generate insoluble, crosslinked fibrin strands, which bind and stabilize weak platelet hemostatic plugs, formed at sites of tissue injury. The formation of this provisional clot is critically dependent on the action of thrombin, and is generated following the stepwise activation of coagulation proteinases via the extrinsic and intrinsic systems. Under normal circumstances, blood is not exposed to tissue factor (TF). However, upon tissue injury, exposure of plasma to TF expressed on non-vascular cells or on activated endothelial cells results in the formation of the TF-activated factor VII (FVIIa) complex. The TF–FVIIa complex subsequently catalyses the initial activation of FX to activated factor X (FXa) and FIX to activated factor IX. FXa in association with activated factor V catalyses the conversion of prothrombin to thrombin. Sustained coagulation is achieved when thrombin synthesized through the initial TF–FVIIa–FXa complex catalyses the activation of FXI, FIX, FVIII and FX. In this manner, the intrinsic pathway is activated.
The systemic inflammatory response syndrome (SIRS) is the massive inflammatory reaction resulting from systemic mediator release that may lead to multiple organ dysfunction. I introduce an analysis of the roles of cytokines, cytokine production, and the relationship of cytokine production to the development of SIRS. The article postulates a three-stage development of SIRS, in which stage 1 is a local production of cytokines in response to an injury or infection. Stage 2 is the protective release of a small amount of cytokines into the body’s circulation. Stage 3 is the massive systemic reaction where cytokines turn destructive by compromising the integrity of the capillary walls and flooding end organs. While cytokines are generally viewed as a destructive development in the patient that generally leads to multiple organ dysfunction, cytokines also protect the body when localized. It will be necessary to study the positive effects of cytokines while also studying their role in causing SIRS. It will also be important to investigate the relationship between cytokines and their blockers in SIRS.
Monocyte/macrophage- and neutrophil-mediated inflammatory responses can be stimulated through a variety of receptors, including G protein-linked 7-transmembrane receptors (e.g., FPR1; MIM 136537), Fc receptors (see MIM 146790), CD14 (MIM 158120) and Toll-like receptors (e.g., TLR4; MIM 603030), and cytokine receptors (e.g., IFNGR1; MIM 107470). Engagement of these receptors can also prime myeloid cells to respond to other stimuli. Myeloid cells express receptors belonging to the Ig superfamily, such as TREM1, or to the C-type lectin superfamily. Depending on their transmembrane and cytoplasmic sequence structure, these receptors have either activating (e.g., KIR2DS1; MIM 604952) or inhibitory functions (e.g., KIR2DL1; MIM 604936).[supplied by OMIM].
TREM-1 associates with and signals via the adapter protein 12DAP12/12TYROBP, which contains an ITAM. To mediate activation, TREM-1 associates with the transmembrane adapter molecule 12DAP12. In sharp contrast to the effect by Ad-FDAP12, transgene expression in the liver of soluble form of extracellular domain of TREM-1 as an antagonist of 12DAP12 signaling, remarkably inhibited zymosan A-induced granuloma formation at every time point examined.
For signal transduction, 01TREM-1 couples to the ITAM-containing adapter DNAX activation protein of 12 kDa (23DAP12 ). MARV and EBOV activate TREM-1 on human neutrophils, resulting in 12DAP12 phosphorylation, TREM-1 shedding, mobilization of intracellular calcium, secretion of proinflammatory cytokines, and phenotypic changes. TREM-1 is the best-characterized member of a growing family of 12DAP12-associated receptors that regulate the function of myeloid cells in innate and adaptive responses. TREM-1 (triggering receptor expressed on myeloid cells), a recently discovered receptor of the immunoglobulin superfamily, activates neutrophils and monocytes/macrophages by signaling through the adapter protein 12DAP12. 522Granulocyte TREM-1 expression was high at baseline and immediately down-regulated upon LPS exposure along with an increase in soluble TREM-1.
DIC is primarily a laboratory diagnosis, based on the combination of elevated fibrin-related markers (FRM), with decreased procoagulant factors and platelets. Non-overt DIC is observed in most patients with sepsis, whereas overt DIC is less frequent. Consumption coagulopathy is a bleeding disorder caused by low levels of platelets and procoagulant factors associated with massive coagulation activation. Treatment with drotrecogin alfa (activated) improves survival and other outcome parameters in severe sepsis, including a subgroup of patients fulfilling the laboratory criteria of overt DIC. No randomized trials demonstrating effective therapies in consumption coagulopathy have been published.
Sepsis is a complex syndrome characterized by simultaneous activation of inflammation and coagulation manifested as systemic inflammatory response syndrome (SIRS)/sepsis symptoms through release of proinflammatory cytokines, procoagulants, and adhesion molecules from immune cells and/or damaged endothelium. Conventional treatments have focused on source control, antimicrobials, vasopressors, and fluid resuscitation; however, a new treatment paradigm exists: that of treating the host response to infection with adjunct therapies including early goal-directed therapy, drotrecogin alfa (activated), and immunonutrition. The drotrecogin alfa (activated) has been shown to reduce mortality in the severely septic patient when combined with traditional treatment. Therapies targeting improved oxygen and blood flow and reduction of apoptosis and free radicals are under investigation. Ultimately, intervention timing may be the most important factor in reducing severe sepsis mortality.
Cell Signaling in Sepsis
Recent data have shown stable patterns of activation among peripheral blood mononuclear cells and neutrophils in healthy human subjects. Although polymorphisms in Toll-like receptors play a contributory role in determining cellular activation, other factors are involved as well. In addition, circulating and locally released mediators of inflammation, including cytokines, complement fragments, and components of activated coagulation and fibrinolytic systems, that are generated in increased amounts during severe infection also interact with membrane-based receptors, leading to activation of intracellular path ways capable of further accelerating proinflammatory cascades. Circulating and organ-specific cell populations are activated to produce proinflammatory mediators during sepsis. Neutrophils and PBMCs bear TLR2 and TLR4, as well as other receptors, such as protein —coupled receptor, that induce increased generation of cytokines and other immunoregulatory proteins, as well as enhance release of proinflammatory mediators, including reactive oxygen species.
The expression of cytokines such as TNF-α and IL-1β is increased in sepsis, and engagement of TNF-α with type I(p55) and type II(p75) TNF receptors or IL-1β with IL-1 receptors belonging to the TLR/IL-1 receptor family produces activation of kinases (including Src, p38, extracellular signal—regulated kinase, and phosphoinositide 3–kinase) and transcriptional factors (such as nuclear factor [NF]–κB) important for further up-regulation of inflammatory proteins.
Genetic polymorphisms lead to alterations in TLR conformation (a small percentage of the variability in humans when their cells are exposed to bacterial products) that are accompanied by decreased cellular activation after exposure to bacterial products. The stable variability in cellular activation that is present among the genetically heterogeneous human population, only a limited number of studies have examined how such patterns may correlate with clinical outcome. A number of studies have examined the transcriptional factor NF-κB and kinases, including p38 and Akt, and provide insights into how heterogeneity in cell signaling may contribute to subsequent clinical course.
Increased activation of the mitogen-activated protein kinase protein 38, Akt, and nuclear factor (NF)–κB in neutrophils and other cell populations obtained at early time points in the clinical course of sepsis-induced acute lung injury or after accidental trauma is associated with a more-severe clinical course, suggesting that a proinflammatory cellular phenotype contributes to organ system dysfunction in such settings. Identification of patients with cellular phenotypes characterized by increased activation of NF-κB, Akt, and protein 38, as well as discrete patterns of gene activation, may permit identification of patients with sepsis who are likely to have a worse clinical outcome, thereby permitting early institution of therapies that modulate deleterious signaling pathways before organ system dysfunction develops, reducing morbidity and improving survival.
NF-kB
The transcriptional regulatory factor NF-κB is a central participant in modulating the expression of many immuno regulatory mediators involved in the acute inflammatory response [30–35]. NF-κB/rel transcription factors function as dimers held latently in the cytoplasm of cells by inhibitory IκB proteins. Signaling pathways initiated by engagement of TLRs, such as TLR 2 and TLR 4, by microbial products and other inflammatory mediators lead to nuclear accumulation of NF-κB and enhanced transcription of genes responsible for the expression of cytokines, chemokines, adhesion molecules, and other mediators of the inflammatory response associated with infection. Association of NF-κB with the inhibitory protein κB-α in the cytoplasm blocks the nuclear localization sequence of NF-κB, inhibiting its movement into the nucleus. Phosphorylation events, in addition to those involving IKKα/β and IκB-α, and involving NF-κB subunits (such as p 65) and nuclear coactivator proteins (such as TATA box binding protein or cAMP-responsive element—binding protein) are mediated by p 38, Akt, and other kinases and play an important role in regulating the transcriptional activity of NF-κB.
Studies have shown that greater nuclear accumulation of NF-κB is accompanied by higher mortality and worse clinical course in patients with sepsis. These clinical series demonstrated that persistent activation of NF-κB was found in nonsurvivors, with surviving patients having lower nuclear concentrations of NF-κB at early time points in their septic course than did nonsurvivors as well as more rapid return of nuclear accumulation of NF-κB. Although studies of patients with sepsis have generally shown that nuclear concentrations of NF-κB are higher in non survivors than in survivors, an unresolved issue is whether such changes occur early and, therefore, define the subsequent course of sepsis or whether pathophysiological changes that result in poor clinical outcome also produce NF-κB activation as a secondary event, so that such changes in NF-κB are simply associated with more severe organ system dysfunction but do not contribute directly to outcome. A study of surgical patients without sepsis supports the hypothesis that neutrophil phenotypes defined by NF-κB activation patterns predict clinical outcome [54]. In that clinical series of patients undergoing repair of aortic aneurysms, higher preoperative levels of NF-κB in peripheral neutrophils were associated with death and with the development of postoperative organ dysfunction.
NF-κB (Photo credit: Wikipedia)
Stable high and low responder phenotypes in the healthy population, implies that the presence of a preexistent high responder neutrophil phenotype, as characterized by increased nuclear translocation of NF-κB after stimulation with TLR 2 or TLR 4 ligands, would be associated with more severe pulmonary inflammatory response and clinical course in response to infection. Conversely, persons whose neutrophils have diminished activation of NF-κB after stimulation would be expected to have less-intense neutrophil-driven inflammation, as well as organ dysfunction. In addition, Nuclear levels of nuclear factor (NF)–κB are significantly increased in neutrophils obtained within 24h of initiation of mechanical ventilation in patients whose clinical course from sepsis-induced acute lung injury is more severe (as defined by death or ventilation for >14 days—that is, ⩽14 ventilator-free days [VFD]), compared with patients with a less-severe course (as defined by mechanical ventilation for <14 days, or >14 VFD). Baseline nuclear concentrations of NF-κB were lower in healthy volunteers than in patients with sepsis-induced acute lung injury, regardless of subsequent clinical course, demonstrating baseline activation of NF-κB in association with sepsis. *P <.05, vs. volunteers. †P< .05, vs. >14VFD.
Modulation of intracellular signaling cascades involving kinases, such as p 38 or Akt, or transcriptional factors, such as NF-κB, through specific inhibitory approaches has shown their pathophysiological importance in experimental models. However, the role of specific intra cellular pathways in contributing to clinical outcomes in patients with sepsis remains incompletely determined, primarily because such alterations in cellular activation patterns have not been examined at early time points before the onset of multiple organ dysfunction. Recent information shows that alterations in p38, Akt, and NF-κB among neutrophils and other cell populations not only precedes the development of organ system dysfunction but also has predictive value in identifying patients with a more severe subsequent clinical course.
Of 289 patients with bacteremia, 52% had normal WBC count and 17% had neither WBC elevation nor fever. Despite multiple studies showing that a normal white blood cell (WBC) count does not exclude serious disease, physicians in all specialties continue to behave as if it did. Among 289 patients (8%) with positive blood cultures, 77% had fever and 48% had elevated WBC count on initial measurement. Neither fever nor an elevated WBC count was noted in 17% of bacteremic patients. The WBC count is not an adequate discriminatory test for bacteremia.
The term SIRS has been widely adopted by clinicians and investigators. As SIRS criteria are not specific for sepsis, several studies have evaluated the usefulness of SIRS criteria for the diagnosis of sepsis. A subanalysis of the Sepsis Occurrence In Acutely Ill Patients (SOAP) study based on intensive care unit (ICU) patients, showed that critically ill patients often fulfill the criteria for SIRS, regardless of whether they are infected or not. At ICU admission, 87% of patients had at least two SIRS criteria, most commonly respiratory rate (84%) or heart rate (71%). There was, however, a higher frequency of three or four SIRS criteria versus two SIRS criteria in infected than in noninfected patients. Interestingly, all infected patients had at least two SIRS criteria. Thus, although the SIRS criteria are so sensitive and are present in so many ICU patients that their value in identifying infected patients is limited, patients fulfilling two or more SIRS criteria should be closely evaluated for infection. The mortality attributable to infection seems to be similar in patients with sepsis and in those without.
The physiologic variables associated with SIRS and sepsis are not only important for the diagnosis and prognosis but also for guiding the treatment. In 1997, the ACCP/SCCM consensus group created a set of practice parameters for the hemodynamic management of patients in septic shock. According to these practice parameters, the initial priority in managing septic shock should be to maintain mean arterial pressure (MAP) by adequate hemodynamic support. In fact, treatment with either MAP or systolic blood pressure as the main therapeutic endpoint is the most commonly recommended and supported clinical monitoring practice. Maintenance of adequate MAP can help ensure adequate organ and tissue perfusion during the time required to detect and treat the infectious process causing the sepsis. the continuous monitoring of physiologic parameters for early sepsis screening continued to center on hypoperfusion and the clinical definitions of hypoperfusion were expanded to include systolic blood pressure less than90 mm Hg, MAP less than 65 mm Hg, a decrease in systolic blood pressure of more than 40 mm Hg, and a decrease in urine output.
This early identification of physiologic instability expedites basic resuscitation. Nine of the 10 signs of vitality can be assessed within several minutes; the 10th [base deficit, calculated after arterial puncture, or central venous oxygen saturation (ScVO2), obtained from a central venous catheter] requires 15-minutes.
ScvO2/base deficit ScvO2<65% or base deficit 5 or lactic acid >2.0 mmol/L
SOFA score is used to predict the probability of infection in critically ill patients based on physiologic parameters like body temperature, respiratory rate, and heart rate, together with other parameters like white blood cell count, C-reactive protein, and Sequential Organ Failure Assessment (SOFA) score by using multiple regression to calculate the relative weight of each parameter in relation to the presence of infection. The significant variables were: body temperature greater than 37.5 C, heart rate greater than 140, and C-reactive protein greater than 6 mg/dL. Among the clinical variables, heart rate was the best predictor of infection, whereas respiratory rate had the poorest predictive value. However, the investigators decided to include all variables in the Infection Probability Score (IPS), assigning points according the relative weight of each parameter in relation to the presence of infection.
A biomarker has been defined as “a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, or pharmacologic responses to therapeutic intervention. The theoretic advantage of combining several biomarkers into a single classification rule is that it should help to improve their classification accuracy and, therefore, their clinical usefulness. when several markers are measured, they are often considered separately, irrespective of the additional information contained in their joint interpretation. This does not constitute a multimarker panel. This distinction is important, because these studies typically describe a list of different biomarkers with potential diagnostic or prognostic values that are used as individual predictors of disease or clinical outcome. Another common confusion is the term multiplex assay, which is a type of laboratory procedure that simultaneously measures multiple analytes (dozens or more) in a single assay. It is distinguished from procedures that measure 1 or a few analytes at a time.9 In contrast, a multimarker panel involves each biomarker representing the inputs to a multivariable, computational prediction or classification model.
Sepsis Bundles
The Institute for Healthcare Improvement (IHI) has highlighted sepsis as an area of focus and has identified several deficiencies that may cause suboptimal care of patients with severe sepsis.
These deficiencies include inconsistency in the early diagnosis of severe sepsis and septic shock, frequent inadequate volume resuscitation without defined endpoints, late or inadequate use of antibiotics, frequent failure to support the cardiac output when depressed, frequent failure to control hyperglycemia adequately, frequent failure to use low tidal volumes and pressures in acute lung injury, and frequent failure to treat adrenal inadequacy in refractory shock.
To address these deficiencies, the Surviving Sepsis Campaign and IHI have revised and added to the Surviving Sepsis Guidelines and created 2 sepsis treatment bundles (resuscitation and management) to guide therapy for patients with severe sepsis.
“Implicit in the use of the bundles is the need to adopt all the elements contained in the bundle,” the authors write. “One cannot choose to apply only selected items from the bundle and expect to achieve comparable benefit. The IHI sepsis website provides tools to screen patients for severe sepsis, as well as to measure success with adherence to implementing the bundles (http://www.ihi.org/IHI/Topics/CriticalCare/Sepsis/).” (The authors are employees of Eli Lilly and Co, the maker of drotrecogin alfa (activated). South Med J. 2007;100:594-600.
The sepsis resuscitation bundle, which should be accomplished as soon as possible and scored during the first 6 hours
Prealbumin (Transthyretin)
Discharge prealbumin and the change in prealbumin were positively correlated with protein and energy intake and inversely correlated with markers of inflammation, particularly CRP and IL-6. When all covariates were included in a multivariable regression analysis, the markers of inflammation predominantly accounted for the variance in prealbumin change (56%), whereas discharge protein intake accounted for 6%.
These authors propose an updated approach that incorporates current understanding of the systemic inflammatory response to help guide assessment, diagnosis, and treatment. An appreciation of a continuum of inflammatory response in relation to malnutrition syndromes is described. This discussion serves to highlight a research agenda to address deficiencies in diagnostics, biomarkers, and therapeutics of inflammation in relation to malnutrition.
Procalcitonin
The most frequent indication for antibiotic prescriptions in the northwestern hemisphere is lower respiratory tract infections (LRTIs),which range in severity from self-limited acute bronchitis to severe acute exacerbation of chronic obstructive pulmonary disease (COPD), and to life-threatening bacterial community-acquired pneumonia (CAP).4 Clinical signs and symptoms, as well as commonly used laboratory markers, are unreliable in distinguishing viral from bacterial LRTI. As many as 75% of patients with LRTI are treated with antibiotics, despitethe predominantly viral origin of their infection. An approach to estimate the probability of bacterial origin in LRTI is the measurement of serum procalcitonin (PCT).
In patients with LRTIs, a strategy of PCT guidance compared with standard guidelines resulted in similar rates of adverse outcomes, as well as lower rates of antibiotic exposure and antibiotic-associated adverse effects. (Trial Registration isrctn.org Identifier: ISRCTN95122877)
Neutrophil CD64
Despite improvements in the treatment of sepsis in recent years, there have been few diagnostic innovations which improve the sensitivity and specificity of diagnosis or facilitate therapeutic monitoring. The clinical reliance on the CBC and leukocyte differential with associated band count to indicate myeloid left shift of immaturity is not accurate, and it is not comparable to the measurement of the metamyeloctes and myelocytes. Only the introduction of a test which measures procalcitonin (PCT), an acute phase marker which is claimed to be more specific for bacterial infections than for viral infections, can be cited as a new diagnostic for the evaluation of patients with suspected infection. A need still persists for improved diagnostic indictors of infection or sepsis, as well as better tests to facilitate monitoring of therapy in the treatment of infection, so that use of antibiotics might be less empirical.
Studies have indicated that quantitative neutrophil CD64 expression is a sensitive and specific laboratory indicator of sepsis or the presence of a systemic acute inflammatory response. Neutrophil CD64 is a highly sensitive marker for neonatal sepsis. Prospective studies incorporating CD64 into a sepsis scoring system are warranted. Studies have indicated that quantitative neutrophil CD64 (high affinity Fc receptor) expression is a worthwhile candidate for evaluation as a more sensitive and specific laboratory indicator of sepsis or the presence of a systemic acute inflammatory response than available diagnostics . Neutrophil (PMN) CD64 is one of many activation-related antigenic changes manifested by neutrophils during the normal pathophysiological acute inflammatory or innate immune response. PMN expression of CD64 is up-regulated under the influence of inflammatory related cytokines such as interleukin 12 (IL-12), interferon gamma (IFN-y) and granulocyte colony stimulating factor (G-CSF).
The first commercially available assay for PMN CD64, developed by Trillium Diagnostics, LLC is a fluorescence based, no wash flow cytometric assay, namely the Leuko64. The assay kit contains a cocktail of monoclonal antibodies including two monoclonal antibodies to CD64 and a monoclonal antibody to CD163, red cell lysis buffer, fluorescence quantitation beads, and a software program for automated analysis of the flow cytometric data that reports PMN CD64 as a CD64 index. The PMN CD64 index is designed so that normal inactivated PMNs yield values of < 1.00 and blood samples from individuals with documented infection or sepsis typically show values > 1.50. Using clinical flow cytometers, the assay can be completed within 30 minutes. While this initial assay format was developed for multiparameter flow cytometers, a new version of the assay has been developed to give nearly identical results on the CD4000 and Sapphire (manufactured by Abbott Diagnostics, Santa Clara, CA) blood cell counters, which are equipped with laser light sources and fluorescence detection capabilities. If these blood cell counters are available in diagnostic haematology laboratories, the Leuko64 assay can be utilised on a 24 hour basis, in contrast to the more typical daytime operation hours of flow cytometric diagnostic laboratories.
Leukocare and Trillium Diagnostics entered an agreement to develop and market Leukocare’s method for detecting inflammatory activity using circulating cell-free DNA. Trillium aims to create a cf-DNA test as a “simple and cost effective” tool that healthcare professionals can use to obtain clinically relevant data on patients who are suspected of having sepsis. The companies said that they expect to finish developing the assay and market it in two years.
B Casserly, R Read, MM Levy. Multimarker Panels in Sepsis. Crit Care Clin 27 (2011) 391–405 doi:10.1016/j.ccc.2010.12.011 criticalcare.theclinics.com
The sepsis consensus conference, led by Roger Bone, promulgated the concept of systemic inflammation as a prime mediator in the “sepsis syndrome”. The members of the consensus committee established “sepsis’ as the intersection between infection and inflammation (defined as SIRS criteria). the greatest importance in defining sepsis lies within the subgroups of the definition: those patients with organ dysfunction (severe sepsis) or refractory hypotension (septic shock). The consensus definition of sepsis emphasized that sepsis may be caused by a variety of infectious organisms (bacteria, fungi, viruses, and parasites) and need not be confined to the bloodstream or even involve bacteremia. Though potentially present in nearly all hospitalized patients, these inclusive criteria set the stage for defining subgroups of sepsis, which have become tremendously important. Septic patients with organ dysfunction were termed “severe sepsis,” while patients with refractory hypotension and/or hypoperfusion were termed “septic shock.”
One failure evident in the current definition is the inability to accurately “stage” the condition of a patient with sepsis. Analogous to staging conducted in the evaluation of malignancy, the experts felt it would be useful to have a similarly predictive process for sepsis. To that end, a new acronym — PIRO — was proposed:
P — Predisposition to sepsis and its altered physiology
I — Infection found to be the cause of sepsis
R — Response to the infection
O — Organ failure
Though vasopressors may be appropriate to increase coronary blood flow in a hypotensive patient with malignant ventricular arrhythmias, the effect of their use on other organ function and even on patient outcome remains uncertain. While we have often strived to maintain normality in critically ill patients, such as arterial blood gases in patient with acute lung injury/acute respiratory distress syndrome, we now realize that such practice is generally incorrect and may have induced harm in the process.
Anti-tumor necrosis factor
Anti-TNF strategies are only partially effective in patients with sepsis. Although individual studies show small, nonsignificant benefits, analysis of all trial data as well as data from a recent trial in a large population of septic patients show that anti-TNF strategies may confer a small survival benefit.
Aprotinin is a serine protease inhibitor that binds to human serine proteases with various affinities. This small protein has profound effects on the systemic inflammatory response syndrome induced by cardiopulmonary bypass. The intersecting pathways of complement, cytokines, coagulation, fibrinolytic, and kinin-kallikrein systems all converge with tremendous amplification and potentially damaging effects to the patient. Following publication of the original study describing high dose aprotinin therapy in repeat coronary artery bypass graft (CABG) surgery in 1987, an extensive body of literature has accumulated supporting the efficacy of aprotinin to reduce blood loss and transfusion requirements in cardiac and orthopedic surgeries. Cardiopulmonary bypass (CPB) initiates the activation of several intersecting plasma protease pathways: the kinin-kallikrein system, the coagulation-fibrinolytic cascade, and the complement system. Activation of each of these pathways results in the production of proinflammatory mediators, resulting in a systemic inflammatory response syndrome (SIRs). Ischemia reperfusion also contributes to the SIRs through the activation of humoral and cellular pathways. Such hemostatic and inflammatory responses are well orchestrated, inter-related processes with the capacity for tremendous amplification.
Activated Protein C
A great advance in treating sepsis patients came with the early completion of the activated protein C trial in June 2000. Since that time, trial results have been peer reviewed and recombinant human activated protein C has become available internationally (drotrecogin alfa [activated]). Recent controversy has arisen regarding protocol changes that occurred during the study and about reduced or absent benefit for patients of the lowest severity of illness. Jay Siegel of the US Food and Drug Administration published the indications for use the New England Journal of Medicine.
The use of activated protein C, as compared with placebo, was associated with a significant reduction in mortality in the Recombinant Human Activated Protein C Worldwide Evaluation in Severe Sepsis (PROWESS) trial (24.7 percent vs. 30.8 percent, P=0.005). Extensive review by physicians and scientists at the FDA confirmed the principal findings of the PROWESS trial but raised issues regarding the interpretation of data and appropriate use of the drug.
These issues were discussed with experts on sepsis from outside the FDA at a public meeting of the agency’s Anti-Infective Drugs Advisory Committee on October 16, 2001. The committee members provided valuable input; highlighted specific issues of concern, some of which are summarized in the accompanying Sounding Board by Warren et al.; and were evenly divided in a vote on a recommendation regarding approval.
The FDA then performed substantial additional evaluations addressing the issues that had been raised and concluded that activated protein C had been demonstrated to be safe and effective in reducing mortality among patients with severe sepsis and a high risk of death, as determined, for example, by the Acute Physiology and Chronic Health Evaluation (APACHE II) score.
Key matters of concern —
changes made during the trial,
the use of APACHE II scores,
and the risk of serious bleeding
In order to increase the capacity of the trial to detect effects on mortality due to sepsis, the eligibility criteria for the activated protein C study attempted to exclude patients who were likely to die from underlying disease within 28 days. The amendments to the eligibility criteria were intended to make this exclusion more effective. Two analyses indicate that these amendments did not account for the improved study outcomes.
Treatment with activated protein C was associated with a 3 percent absolute reduction in mortality among patients without chronic health points on APACHE II but with a 19 percent absolute reduction in mortality among patients who had chronic health points. The P value for the interaction of treatment effect and APACHE II chronic health points was 0.01. Both of these analyses indicate that the amendments to the protocol, rather than accounting for improved results, actually excluded patients who appeared more likely to benefit from therapy. In addition, these two analyses address the generalizability of the results of the PROWESS trial to patients with sepsis who have serious preexisting disease.3 They suggest strongly that serious preexisting disease should not, in general, be a reason for withholding activated protein C therapy.
A1AR Antagonists: Disease Modifying Drug Candidates
Block endotoxin induced acute lung injury
Block ischemia reperfusion injury of lung, heart, and liver
Increase survival and reduce ALI in animal model of intra-abdominal polymicrobial sepsis (rat CLP) (Endacea data; NIH/NIAID-funded)
Increase survival and reduce ALI in rat model of pneumonic plague (Endacea data; NIH/NIAID-funded)
Highly selective for human A1AR, water soluble, orally bioavailable A1AR antagonist
Analog of an approved off-patent anti-asthma drug
L-97-1 is a proprietary analog of bamiphylline with:
Greater affinity for the human A1AR (increased potency)
Greater selectivity for the human A1AR
And L-97-1 retains the water solubility of bamiphylline