Feeds:
Posts
Comments

Posts Tagged ‘bone marrow’

 

Nanoparticles can turn off genes in bone marrow cells

Reporter : Irina Robu, PhD

MIT engineers developed an alternative to turn off specific genes which play a vital role in producing blood cells of the bone marrow using specialized nanoparticles. These nanoparticles can be made-to-order to treat heart disease or increase the yield of stem cells in patience who need stem cell transplants. The particles are coated with lipids that help stabilize them, and they can target organs such as the lungs, heart, and spleen, depending on the particles’ composition and molecular weight. This genetic therapy, also known as RNA interference is difficult to target organs other than the liver, where most of the nanoparticles tend to collect.

RNA interference is an approach that could theoretically be used to treat a variety of diseases by delivering short strands of RNA that block specific genes from being turned on in a cell. Yet, the main obstacle to this kind of therapy has been  delivering it to the right part of the body. When injected into the bloodstream, nanoparticles carrying RNA tend to accrue in the liver, which various biotech companies have taken advantage of to develop new experimental treatments for liver disease.

In their recent study, scientists set out to adapt the nanoparticles so that they could reach the bone marrow which contains stem cells that produce different types of blood cells. Stimulating the process , they could enhance the yield of hematopoietic stem cells for stem cell transplantation and they created variants that have different arrangements of surface coating, polyethylene glycol. They were able to test 15 particles and determined one that was able to avoid being caught in the liver or the lungs, and that could effectively accumulate in endothelial cells of the bone marrow. They also showed that RNA carried by this particle could reduce the expression of a target gene by up to 80 percent.

The scientists then tested this approach with two genes. The first gene, SDF1 is a molecule that normally prevents hematopoietic stem cells from leaving the bone marrow. They realized that turning off the SDF1 gene could have the same effect as the drugs that are being used to induce hematopoietic stem cell release in patients who need undergo radiation treatments for blood cancers. These stem cells are later transplanted to repopulate the patient’s blood cells. By knocking down SDF1, they could boost the release of hematopoietic cells fivefold which is comparable to the levels achieved by the drugs that are now used to enhance stem cell release.

The second gene researchers use is MCP1, a molecule that plays a key role in heart disease.  They realized that when MCP1 is released by bone marrow cells after a heart attack, it stimulates a flood of immune cells to leave the bone marrow and travel to the heart. Researchers realize that by delivering RNA that targets MCP1 reduced the number of immune cells that went to the heart after a heart attack.

Using these new particles, researchers hypothesized that they could further develop treatments for heart disease and other conditions.

SOURCE

https://news.mit.edu/2020/nanoparticles-bone-marrow-rnai-1005

Read Full Post »

Monitoring AML with “cell specific” blood test

Larry H. Bernstein, MD, FCAP, Curator

LPBI

 

‘Liquid Biopsy’ Blood Test Replaces Painful Bone Marrow Biopsy for Leukemia

Mon, 01/11/2016  by BioFluidica, Inc.  http://www.mdtmag.com/news/2016/01/liquid-biopsy-blood-test-replaces-painful-bone-marrow-biopsy-leukemia

 

BioFluidica, Inc. has released the clinical data for minimal residual disease detection in Acute Myeloid Leukemia (AML) patients using circulating leukemic cells selected from blood. The data was published in the peer reviewed journal the Analyst (141 (2016) 640). AML is a rapidly developing leukemic disease with ~20,000 cases reported in 2015 with a 5-year survival rate of only 25%.

The goal of this study was to detect early stages of disease relapse following stem cell transplantation. Currently AML relapse is detected using bone marrow biopsy samples that are painful for the patient and using existing commercial tests, limits the frequency of testing and thus resulting in poor outcomes for AML patients. The paper describes that using BioFluidica’s analytical technology relapse could be detected nearly 2 months earlier than conventional tests. In addition, test frequency could be significantly increased using BioFluidica’s technology compared to tests requiring bone marrow biopsies.

Professor Steven A. Soper, the scientific founder of BioFluidica and co-author of the paper with Dr. Paul Armistead, a hematologist, both at the University of North Carolina states that “the use of a blood test compared to a bone marrow biopsy would be a tremendous advancement in diagnostic capability that can dramatically improve the survival rate of patients with AML.”

BioFluidica is developing innovative technologies for the isolation and analysis of rare, circulating biomarkers in the blood. The company’s first platform has the capacity to isolate circulating tumor cells, exosomes and cfDNA from the blood with unprecedented recovery and purity. The technology is based on patented microfluidics designs which has been clinically validated for 6 different cancer types including Colorectal, Pancreatic Ductal Adenocarcinoma, Ovarian, Breast, Multiple Myeloma and AML. Additionally, stroke detection and infectious disease identification have also obtained clinical validation using the BioFluidica test. The company was cofounded by Dr. Soper who is currently a Professor in Biomedical Engineering and Chemistry at the University of North Carolina at Chapel Hill (UNC-CH). He is also Director of a new center on the UNC-CH campus, Center for BioModular Multi-scale Systems for Precision Medicine, focused on developing new tools for the molecular analysis of circulating biomarkers.

Read Full Post »

Reporter: Aviva Lev-Ari, PhD, RN

The Bone Marrow Niche, Stem Cells, and Leukemia: Impact of Drugs, Chemicals, and the Environment

May 29 – 31, 2013
The New York Academy of Sciences

Presented by Rutgers, The State University of New Jersey and the New York Academy of Sciences

Register Now

Over 20,000 Americans are diagnosed each year with bone marrow failure syndromes. Environmental, chemical, and genetic factors have been linked to the development of lymphomas, leukemias, and myelodysplastic syndromes (MDS). Additionally, some anti-cancer drugs have been shown to themselves induce DNA damage and secondary cancers. In light of increasing societal exposure to toxic environmental agents that may be carcinogenic, including chemicals and pharmaceuticals, we face the potential for a rise in the incidence of bone marrow failure and malignancy. In order to better understand leukemia it may be necessary to examine it from the perspective that it is an environmental disease.

To date, two separate groups of scientists and physicians have been studying bone marrow: toxicologists who examine the effects of chemicals and the environment on healthy marrow, and hematologists and oncologists who investigate bone marrow abnormalities and malignancies. Thus, there is a clear, unmet need for collaboration between these fields within academia, industry, and government in order to accelerate our investigation and understanding both of basic bone marrow biology and chemically-induced diseases of the marrow.

This 2.5-day conference will bring together representatives from two areas of research, toxicology and hematology, around a jointly shared goal — to better understand, prevent, and treat myeloid neoplasms. Conference Sessions will combine basic science and toxicology research at the level of the bone marrow niche with clinical findings from healthy subjects and patients. Topics for discussion will include bone marrow niche structure and function, the maturation and differentiation of healthy and leukemogenic hematopoietic stem cells, and the environmental, chemical, and genetic factors involved in the development of myeloid abnormalities including MDS and acute myeloid leukemia (AML). The meeting will feature a series of plenary lectures, panel discussions, a poster session, and short talk presentations selected from abstracts submitted by early career investigators.

Organizing Committee*

Conference Organizers

Michael A. Gallo, PhD

Robert Wood Johnson Medical School and Environmental and Occupational Health Sciences Institute; Rutgers, The State University of New Jersey

Helmut Greim, MD

Technical University of Munich

Robert Snyder, PhD (Chair)

Environmental and Occupational Health Sciences Institute; Rutgers, The State University of New Jersey

Subcommittee Chairs

Finance:

Robert Snyder, PhD

Environmental and Occupational Health Sciences Institute; Rutgers, The State University of New Jersey

International Advisory Committee:

Helmut Greim, MD

Technical University of Munich

Logistics:

Debra Kaden, PhD

Environ International Corporation

Programs:

Richard Larson, MD

University of Chicago

David Ross, PhD

University of Colorado Anschutz Medical Campus

Publications:

Jerry M. Rice, PhD

Georgetown University Medical Center

* Please click on the Speakers tab for a complete listing of the Organizing Committee

Registration Pricing

By 4/26/2013 After 4/26/2013 Onsite
Member $350 $400 $500
Student/Postdoc Member $200 $250 $300
Nonmember (Academia) $400 $450 $550
Nonmember (Corporate) $500 $550 $650
Nonmember (Non-profit) $400 $450 $550
Nonmember (Student / Postdoc / Fellow) $200 $250 $300

Registration includes a complimentary, one-year membership to the New York Academy of Sciences. Complimentary memberships are provided to non-members only and cannot be used to renew or extend existing or expiring memberships. A welcome email will be sent upon registration which will include your membership credentials.

Presented by

  • The New York Academy of Sciences
  • Rutgers University

http://www.nyas.org/Events/Detail.aspx?cid=b4c60844-7a53-45d6-8640-9419c6353529

Read Full Post »

Positioning a Therapeutic Concept for Endogenous Augmentation of cEPCs — Therapeutic Indications for Macrovascular Disease: Coronary, Cerebrovascular and Peripheral

Author and Investigator Initiated Study: Aviva Lev-Ari, PhD, RN

 

Macrovascular Disease – Therapeutic Potential of cEPCs: Promise for CV Risk Reduction

  • Introduction
  • Biomarker Discovery – a comprehensive Post on this topic is forthcoming
  • What are our Contributions in the Domain of Macrovascular Disease – Therapeutic Potential of cEPCs: Reduction Methods for CV Risk
  • Postulates of Multiple Indications for the Method Presented: Positioning of a Therapeutic Concept for Endogenous Augmentation of cEPCs — Potential Therapeutic Indications for ElectEagle
  • A Three Component Method for Endogenous Augmentation of cEPCs — Macrovascular Diseases – Therapeutic Potential of cEPCs
  • The Promise of the Proposed Pharmacotherapy as a Method of CVD Risk Reduction
  • Emergence of Clinical Trial Results on Genous R stent — Healthy Endothelial Accelerated Lining Inhibits Neointimal Growth – (HEALING II)
  • Conclusions
  • References

Key words: coronary artery disease, circulating Endothelial Progenitor Cells (cEPCs), Endothelial Progenitor Cells (EPC), genetic engineering, CVD, CAD, CHF, myocardial infarction, neovascularization, vascular repair, “multimarker biomarker”, macrovascular disease, Endogenous Augmentation of cEPCs, Primary Endpoint, Secondary Endpoint.

Abbreviations used: ED, endothelial dysfunction; CAD, coronary artery disease; CVD, cardiovascular disease; cEPCs, circulating Endothelial Progenitor Cells; EPC, Endothelial Progenitor Cells; CHF, congestive heart failure; MI, myocardial infarction; MNC, mononuclear cells; VEGF, vascular endothelial growth factor; BMMNCs, bone marrow-derived mononuclear cells; G-CSF, granulocyte colony-stimulating factor; SDF, stromal derived factor; PB-MNCs, peripheral blood-mononuclear cells; EF, ejection fraction; PO2, partial pressure of oxygen; BMS, bare-metal stent; CABG, coronary artery bypass graft; DES, drug-eluting stent; GP, glycoprotein; LAD, left anterior descending; LCx, left circumflex; MI, myocardial infarction; RCA, right coronary artery; S/P , status-post stent implantation; MACE, Major Adverse Cardiac Events; TLR, target lesion revascularization; TVR, target vessel revascularization; TVF, target lesion vessel failure; eNOS, endothelial Nitric Oxide Synthase 

Introduction

Cardinal to the study of reendothelialization and neovascularization is the mechanism of action (MOA) of EPCs. It requires exact biological phenotype of the true EPC and its MOA on the endothelium. Is the EPC autocrine or paracrine in its functional role? It is critical to understand this biological unknown for planning therapeutic approaches. Patients with unstable angina and no evidence of cardiac necrosis exhibited increased cEPCs. Systemic inflammation and recognized growth factors may play a role in peripheral mobilization of EPCs in patients with unstable anginal syndromes. Proportion of cEPCs in coronary ischemia, acute or chronic and its potential for restoring left ventricular dysfunction is still experimental. EC injury facilitates an accelerated development of atherosclerotic plaque which triggers cardiovascular risk factors where the magnitude of the endothelial dysfunction predicts the level of risk for a macrovascular event (George, 2004).

Diminished level of cEPCs is associated with risk factors for CVD implicating impaired endothelial repair as a contributor to a dynamic state of endothelial dysfunction. cEPCs is further reduced if multiple risk factors for CVD are present. Endothelial dysfunction is associated with cEPCs counts. It is only if cEPCs counts are low then endothelial dysfunction (ED) emerges. In the case of ED, the cells were more senescent compared with an age group without CVD and the risk factors involved with it. Impaired repair capacity due to reduced availability of cEPCs enhances the exposure to risk factors when injury occurs due to endothelial denudation, ischemic tissue, neointima build up and remodeling.

Mobilization and EPC-mediated neovascularization is critically regulated. Statins and physical exercise are stimulatory while risk factors for CAD are inhibitory in the modulation function of the level of cEPCs. Recruitment of cEPCs requires a coordinated sequence of adhesive and signaling events including adhesion and migration by integrins, chemoattraction of SDF-1/CXCR4 and differentiation of EC.

Bone-marrow derived cells in the circulating blood have an endothelial phenotype and peripheral blood can be cultured to generate ECs. cEPCs provide both diagnostic and prognostic information on CVD. EPCs are analyzed by their phenotypic markers, as discerned by fluorescence-activated cell sorting (FACS) analysis as well as by their functional capability to produce colonies in culture conditions.

Kiernan (2006) identifies the two classes of therapeutic applications of cEPCs: (a) induction of angiogenesis and (b) large vessel repair. Transplantation of autologous EPCs over-expressing eNOS in injured vessels enhances the vasculoprotective properties of the reconstituted endothelium, leading to inhibition of neointimal hyperplasia. This cell-based gene therapy strategy may be useful in treatment of vascular disease. Stents coated in CD34 antibody which binds to the CD34 antigen of cEPCs have the capability to promote re-endothelialisation in minutes to hours. This mechanism seeks to restore the normal biology of the vessel wall rather than perpetuate the wall disruption as drug eluting stents are found recently to be implicated to cause both restenosis and thrombosis (Tung et al., 2006). Thus, cEPCs are of cardinal importance in healing cardiovascular injury. Identification of augmentation methods which are endogenous in nature, are systemic rather than local, as cell-based therapy is, and therefore, it will deliver systemic protective measures against atherosclerosis delaying angioplasty and potentially avoiding cell implantation or vascular engrafting.

Biomarker Discovery – a comprehensive Post on this topic is forthcoming

A comprehensive review of “Traditional” vs. “Novel” risk markers for cardiovascular disease was recently undertaken by Folsom et al., (2006) and the Editorial to this article by Lloyd-Jones and Tian (2006). Among the “Traditional” Risk Markers, they list: Age, Race, Sex, Total/HDL levels, Smoking Status, Diabetes, Systolic BP and Use of antihypertensive  drugs. The list of “Novel” Risk Markers is impressively longer and includes: CRP, Lp-PLA2, E-Selectin, Fibrinogen, PAI-1, Vitamin B6, D-dimer, ICAM-1, Homocysteine, IL-6, HSV-1 Antibody, CMV Antibody and Folate.

Only two risk factors make the top five list following the data adjustment to Age and /or All the Traditional Risk Factors, respectively, I would conclude that only the following two are of paramount importance for clinical application and drug therapy design.

Risk Factor RANKING

Risk Factor RANKING if

Data Adjusted to

AGE

Risk Factor RANKING if

Data Adjusted to

All “Traditional” Risk Factors

1 Chlamydia Intracellular adhesion molecule
2 Lp-PLA2 lipoprotein-associatedphospholipase A2 Cytomegalovirus
3 Tisshe Plasminogen Activator D-Dimer
4 Tissue inhibitor of Metalloproteinase1 IL-6
5 Intracellular adhesion molecule Tissue inhibitor of Metalloproteinase1

In light of these results, chiefly edified by Folsom et al., (2006)  conclusion that: “Based on the totality of evidence, however, CRP level does not emerge as a clinically useful addition to basic risk factor assessment for identifying patients at risk of a first CHD event.” (Folsom, 2006, 1372).

What are our Contributions in the Domain of

Macrovascular Disease – Therapeutic Potential of cEPCs: Reduction Methods for CV Risk

(a) This is the first paper to look at cEPCs from two academic schools of thought.  One, represented by the review article of Dzau et al., Hypertension, 2005 with 122 references which treats cEPCs from two perspectives: Vascular Biology and Molecular Cardiology. The other, is the review article by Lapidot & Petit, Experimental Hematology, 2002 with 86 references which treats cEPCs as stem cells and covers the research in Immunology and in Hematology, cEPCs is circulating in our blood, it is a stem cell! The overlap between the references N=122 in Dzau and N=86 in Lapidot & Petit is zero. These two schools do not cite the findings of the other school. That happens when both schools (Vascular Biology/Molecular Cardiology) and (Immunology/Hematology), BOTH schools are researching the same biologic phenomenon, i.e., one circulating EPC. We are the first to put together in one paper the two schools in the context of cEPCs. The pathophysiology of cECs, cEPCs and Trans-Endothelium Cell Migration in one location.

(b) Table of content of Part I yielded a theoretical treatment of cEPCs not in existence anywhere.  We defined for the first time that the Clinical Frontier for cEPCs is of quadruple nature: (Vascular Biology/Molecular Cardiology) PLUS (Immunology/Hematology). We made the statement that the Clinical Frontier has 20 Future Fast Acting Therapy modality currently under research. We cited the limitation of exogenous methods for augmentation of cEPCs as a scientifically derived justification for our selection of an endogenous augmentation method.

Upon selection of the endogenous method, we specified three components:

–   inhibition of ET-1

–   induction of eNOS

–   stimulation of PPAR-gamma

The proposed combination drug therapy yielded a new multimarker biomarker for reduction of CVD risk for macrovascular events, called the ElectEagle Version I. We specified the potential indications for the ElectEagle Version I method in terms of cardiovascular disease and co-morbidity with other endothelial dysfunction derived disease.

Method name:            ElectEagle

E.L.E.C.T.

E – Efficient

L – Ligands of cEPCs

E – Elective and Individualized Diagnosis and Therapy

C – Cardiovascular diseases & secondary sequalea

T – Treatment adjustable by three agents

E.A.G.L.E.

E – Endogenous

A – Augmentation

G – Gamma-PPAReceptor

L – Ligand occupied ETA and ETA-ETB – binding Nitric Oxide

E – EPCs fast generator

ElectEaglestands for an Efficient Ligands of cEPCs Elective and Individualized Diagnosis and Therapy for Cardiovascular diseases & secondary vascular sequalea, using Treatment adjustable by three agents. It is a method for Endogenous Augmentation of circulating EPCs by using Gamma-PPAR agonists, inhibitors of Ligand occupied ETA and ETA-ETB and agonist for binding Nitric Oxide and induce eNOS.

A Three Component Method for Endogenous Augmentation of cEPCs — Macrovascular Diseases – Therapeutic Potential of cEPCs

Observations on Intellectual Property Development For an Unrecognized Future Fast Acting Therapy for Patients at High Risk for Macrovascular events

ElectEagle represents a discovery of a novel “multimarker biomarker” for cardiovascular disease that innovates on four counts.

First, it proposes new therapeutic indications for acceptable drugs.

Second, it defines a specific combination of therapeutic agents, thus, it put forth a proprietary drug combination.

Third, it targets receptor systems that have not been addressed in the context of cEPCs augmentation methods. Chiefly, modulation of the following three-targeted receptor systems: (a) inhibition of ET-1, ETA and ETA-ETB receptors by antagonists (b) induction of eNOS, by agonists and NO stimulation and (c) upregulation of PPAReceptor-gamma by agonists (TZD). While (b) and (c) are implicated as having favorable effects of cEPCs count, each exerting its effect by a different pathway, it is suggested in this project that (a) might be identify to be the more powerful of the three markers. Our method, ElectEagleis the FIRST to postulate the following: (1) time concentration dependence on eNOS reuptake (2) dose concentration dependence on NO production (3) time and dose concentration dependence for ET-1, ETA and ETA-ETB inhibition, and (4) dose concentration dependence on PPAReceptor-gamma. Points First, Second and Third are covered in Part II where a special focus is placed on ET-1, ETA and ETA-ETB receptors.

Fourth, ElectEagle proposes a platform with triple modes of delivery and use of the test, as described in Part III. The triple modes are as follows: (A) an automated platform from a centralized lab with integration to Lab’s information management system. (B) a point-of-care testing device with appropriate display of test results (small benchtop analyzers in PCP office). (C) a device used for home monitoring of analytes (the hand-held device facilitates rapid read of scores and their translation to drug concentration of each of the three therapeutic agents, with computation of the three drug concentrations done by the device. Thus, it offers quicker optimization of treatment.  ElectEagle is the FIRST to propose a CVD patient kit, hand-held device, which calculates on demand an adjustable therapeutic regimen as a function of cEPCs count biomarker. In this regard, a similarity to the pump, in management of blood sugar in DM patients, exists. Since there is a high co-morbidity between DM and CVD, our methods, ElectEagle may eventually become a targeted therapy for the DM Type 2 population.

Postulates of Multiple Indications for the Method Presented: Positioning of a Therapeutic Concept for Endogenous Augmentation of cEPCs — Potential Therapeutic Indications for ElectEagle

ElectEagle can become the drug therapy of choice for the following indications:

  •      CAD patients
  •      Endothelial Dysfunction in DM patients with or without Erectile Dysfunction
  •      Atherosclerosis patients: Arteries and or veins
  •      pre-stenting treatment phase
  •      post-stenting treatment phase
  •      if stent is a Bare Metal stent (BMS)
  •      if stent is Drug Eluting stent (DES)
  •      if stent is EPC antibody coated (the ElectEagle method increase cEPCs generation in vitro) so availability of cEPCs is increased
  •      post CABG patients (the ElectEagle enhances healing by endogenous augmentation of cEPCs)
  •      target sub segments of CVD patients on blood thinner drugs (the ElectEagle does not require treatment with antiplatelet agents, it is suitable for all patients on Coumadin. This population have a counter indication for antiplatelet agents which is a follow up treatment after stent implantation for 30 days, with stent-eluting long term regimen of antiplatelet agents, 6 months and in some cases indefinitely (Tung, 2006).
  •      ElectEagle is based on systemic therapeutics (versus the localized stent solution requiring multiple and even overlapping stents)
  •      ElectEagle will be having potential in three contexts

(a) Coronary disease

(b) Periphery vascular disease

(c) Cerebrovascular

Comparative analysis of endogenous and exogenous cEPCs augmentation methods:

A. Endogenous augmentation method properties:

  •         temporal – while drug therapy in use – drug action is interruptible
  •         time concentration on eNOS reuptake
  •         dose concentration on NO production
  •         time and dose concentration manner for ETB inhibition
  •         dose concentration on PPAR-gamma

B.  Cell-based and other exogenous methods

  • permanent colonization till apoptosis if no repeated attempts of re-transfer,
  • re-implantation as the protocol usually has several stages

The Promise of the Proposed Pharmacotherapy as a Method of CVD Risk Reduction

It is expected that ElectEagle will be resulting in potential delay of stenting implantation. Patients that are target for stenting may benefit form ElectEagle that will facilitate and accelerate healing after the stent is in place. EPC antibody coated stents will work if and only if the patient has more that just low cEPCs, most patient undergoing stenting tend to have low level of cEPC. The ElectEagle method can be coupled with that type of new stents, called Genous, now in clinical trials (HEALING II, III). These stents enhance the body ability in mobilization of cEPCs, only. However, if the initial population of cEPCs is low, an endogenous fast acting cell augmentation method is needed for pretreatment before the PCI procedure with Genous is scheduled.

Emergence of Clinical Trial Results on Genous R stent — Healthy Endothelial Accelerated Lining Inhibits Neointimal Growth – (HEALING II)

Latest publications on HEALING II – Clinical Trial of EPC coated stent

Genous R stent
n=63
Healthy Endothelial Accelerated Lining Inhibits Neointimal Growth – II

S Silber et al; 12 Month Outcomes of the e-HEALING (Healthy Endothelial Accelerated Lining Inhibits Neointimal Growth) Worldwide Registry; EuroIntervention 2011;6:819-825

P Damman et al; Coronary Stenting With the Genous Bio-engineered R stent in Elderly Patients – 12-month Outcomes From the e-HEALING Registry; Circulation Journal 2011;75(11):2590-2597

P Damman et al; Twelve-month Outcomes After Coronary Stenting With the Genous Bio-Engineered R Stent in Diabetic Patients from the e-HEALING Registry; Journal of Interventional Cardiology 2011;24(4):285-94 

J Aoki et al; Endothelial progenitor cell capture by stents coated with antibody against CD34: the HEALING-FIM (Healthy Endothelial Accelerated Lining Inhibits Neointimal Growth-First In Man) Registry.J.Am.Coll.Cardiol. 2005 May 17;45(10):1574-9

REFERENCES

Aicher A, Heeschen C, Mildner-Rihm C, Urbich C, Ihling C, Technau- Ihling K, Zeiher AM, Dimmeler S, (2003). Essential role of endothelial nitric oxide synthase for mobilization of stem cell and progenitor cells. Nat Med., 9:1370-1376.

Anderson T. (1999). Assessment of treatment of endothelial dysfunction. J Am Coll of Cardiology, 34: 631- 8.

Andrew C. Li, Binder, CJ, Gutierrez, A, Brown, KK, Plotkin, CR, Pattison, JW, Valledor, AF, Davis, RA, Willson, TM, Witztum, JL, Palinski, W, Glass, CK. (2004). Differential inhibition of macrophage foam-cell formation and atherosclerosis in mice by PPAR-alpha, Beta/delta, and gamma. J. Clin. Invest. 114:1564-1576

http://www.jci.org/articles/view/18730

Aoki, J., Serruys, P.W., van Beusekom, H., Ong, A.T., McFadden, E.P., Sianos, G., et al. (2005). Endothelial progenitor cell capture by stents coated with antibody against CD34: the HEALING-FIM (Healthy Endothelial Accelerated Lining Inhibits Neointimal Growth-First In Man) Registry. J Am Coll Cardiol 45 (10), 1574–1579.

Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatterman G, and Isner JM (1997). Isolation of putative progenitor endothelial cells for angiogenesis. Science 275: 964–967.

Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, Kearne M, Magner M, Isner JM. (1999). Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res. 85:221–228.

Askari AT, Unzek S, Popovic ZB, Goldman CK, Forudi F, Kiedrowski M, Rovner A, Ellis SG, Thomas JD, DiCorleto PE, Topol EJ, Penn MS.(2003). Effect of stromal cell-derived factor 1 on stem cell homing and tissue regeneration in ischemic cardiomyopathy. Lancet, 362:697–703.

Assmus B, Schachlinger V, Teupe C, Britten M, Lehmann R, Dobert N, Grunwald F, Aicher A, Urbich C, Martin H, Hoelzer D, Dimmeler S, Zeiher AM, (2002). Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (TOPCARE-AMI). Circulation, 106:3009 –3017

Bennett MR, O’Sullivan MO (2001). Mechanisms of angioplasty and stent restenosis: implications for design of rational therapy. Pharmacol Ther., 91:149 –166.

Ben-Shoshan, J and George, J. (2006). Endothelial progenitor cells as therapeutic vectors in cardiovascular disorders: from experimental models to human trials  Pharmacology Therapeutics (impact factor: 8.9). 08/2007; 115(1):25-36.

Bhattacharya V, Shi Q, Ishida A, Sauvage LR, Hammond WP, Wu MH.(2000). Administration of granulocyte colony-stimulating factor enhances endothelialization and microvessel formation in small caliber synthetic vascular grafts. J Vasc Surg., 32:116 –123.

Bonetti PO, et al. (2002). Reactive hyperemia peripheral arterial tonometry, a novel non-invasive index of peripheral vascular function, is attenuated in patients with coronary endothelial dysfunction. Circulation, 106:Suppl II:579.

Bonetti PO, et al. (2003). Enhanced external counterpulsation improves endothelial function in patients with symptomatic coronary artery disease. J Am Coll of Cardiology, 41:1761-8.

Britten MB, Abolmaali ND, Assmus B, Lehman R, Honold J, Schmitt J, Vogl TJ, Martin H, Schachinger V, Dimmeler S, Zeiher AM, (2003). Infarct remodeling after intracoronary progenitor cell treatment in patients with acute myocardial infarction (TOPCARE-AMI): mechanistic insights from serial contrast-enhanced magnetic resonance imaging. Circulation, 108:2212–2218.

Bypass Angioplasty Revascularization Investigation in Type 2 Diabetics (BARI 2D) ClinicalTrials.gov Identifier: NCT00006305, 2000-2007

http://www.nejm.org/doi/full/10.1056/NEJMoa0805796

Caplice NM, Bunch TJ, Stalboerger PG, Wang S, Simper D, Miller DV, Russell SJ, Litzow MR, Edwards WD. (2003). Smooth muscle cells in human coronary atherosclerosis can originate from cells administered at marrow transplantation. Proc Natl Acad Sci U S A. 100: 4754–4759.

Chadwick , D.(2006) OrbusNeich’s Genous Bioengineered R-stent . Cath Lab Digest, 14 (1), 20-26

Cho H-J, Kim H-S, Lee M-M, Kim D-H, Yang H-J, Hur J, Hwang K-K, Oh S, Choi Y-J, Chae I-H, Oh, B-H, Choi Y-S, Walsh K, Park Y-B. (2003).  Mobilized endothelial progenitor cells by granulocyte-macrophage colony-stimulating factor accelerate reendothelialization and reduce vascular inflammation after intravascular radiation. Circulation, 108:2918 –2925.

Choi J-H, Kim KL, Huh W, Kim B, Byun J, Suh W, Sung J, Jeon E-S, Oh H-Y, Kim D-K, (2004). Decreased number and impaired angiogenic function of endothelial progenitors in patients with chronic renal failure. Arterioscler Thromb Vasc Biol.,24:1246 –1252.

Cinamon G, Shinder V, Alon R (2001) Shear forces promote lymphocyte migration across vascular endothelium bearing apical chemokines. Nature Immunology, 2:515

Dimmeler S, Aicher A, Vasa M, Mildner-Rihm C, Adler K, Tiemann M, Rutten H, Fichtlscherer S, Martin H, Zeiher AM, (2001). HMG-CoA reductase inhibitors (statins) increase endothelial progenitor cells via the PI3-kinase/Akt pathway. J Clin Invest., 108:391–397.

Dimmeler S and Zeiher AM, (2004). Vascular repair by circulating endothelial progenitor cells: the missing link in atherosclerosis. J Mol Med. 82:671– 677.

Drexler H and Hornig B, (1999). Endothelial dysfunction in human disease. J Mol Cell Cardiol., 31:51– 60.

Dzau VJ, Braun-Dullaeus RC, Sedding DG. (2002). Vascular proliferation and atherosclerosis: new perspectives and therapeutic strategies. Nat Med.,  8:1249 –1256.

Dzau, VJ, Gnecchi, M, Pachori, AS, Morello F, Melo, LG.(2005).Therapeutic Potential of Endothelial Progenitor Cells in Cardiovascular Diseases.Hypertension,246:7-18.

Edelberg JM, Tang L, Hattori K, Lyden D, Rafii. (2002). Young adult bone marrow-derived endothelial precursor cells restore aging-impaired cardiac angiogenic function. Circ Res., 90:e89–e93.

Endemann DH and Schiffrin EL, (2004). Endothelial dysfunction. J Am Soc Nephrol., 15:1983–1992.

Fadini, G, Avogaro A, Agostini C. (2004), Unambiguous Definition of Endothelial Progenitor Cells. Electronic Letter to the Editor of Heart in reference to article by Eizawa, T, Ikeda U, et al. Decreasing in circulating endothelial progenitor cells in patients with stable CAD, Heart, 2004; 90: 685-686,

http://heart.bmjjournals.com/cgi/eletters/90/6/685#310  retrieved on 6/26/2006 Link not found 5/1/2013

Fernandez-Aviles F, San Roman JA, Garcıa-Frade J, Fernandez ME, Penarrubia MJ, de la Fuente Luis, Gomez-Bueno M, Cantalapiedra A, Fernandez J, Gutierrez O, Sanchez PL, Hernandez C, Sanz R, Garcıa- Sancho J, Sa´nchez A, (2004). Experimental and clinical regenerative capability of human bone marrow cells after myocardial infarction. Circ Res., 95:742–748.

Folsom, A.R. Chambless, L.E. Ballantyne, C.M. Coresh, J. Heiss, G. Wu, K.K. Boerwinkle, E. Mosley, T.H. Sorlie, P. Diao, G. Sharrett, A.R. (2006). An Assessment of Incremental Coronary Risk Prediction Using C-Reactive Protein and Other Novel Risk Markers – The Atherosclerosis Risk in Communities Study. Arch Intern. Med. 166, 1368-1373.

Fuujiyama S, Amano K, Uehira K, Yoshida N, Nishiwaki Y, Nozawa Y,  Jin D, Takai S, Miyazaki M, Egashira K, Imada T, Iwasaka T, Matsubara H, (2003). Bone marrow monocyte lineage cells adhere on injured endothelium in a monocyte chemoattractant protein-1-dependent manner and accelerate reendothelialization as endothelial progenitor cells. Circ Res., 93:980-989.

George F, Brisson C, Poncelet P, Laurent JC, Massot O, Arnoux D, Ambrosi P, Klein-Soyer C, Cazenave JP, and Sampol J (1992). Rapid isolation of human endothelial cells from whole blood using S-Endo 1 monoclonal antibody coupled to immunomagnetic beads: demonstration of endothelial injury after angioplasty.Thromb Haemost, 67:147–153.

George J, Herz I, Goldstein E, Abashidze S, Deutch V, Finkelstein A, Michowitz Y, Miller H, Keren G.(2003). Number and adhesive properties of circulating endothelial progenitor cells in patients with in-stent restenosis. Arterioscler Thromb Vasc Biol., 23:e57– e60.

George J, Goldstein E, Abashidze S, Deutsch V, Shmilovich H, Finkelstein A, Herz I, Miller H, Keren G., (2004). Circulating endothelial progenitor cells in patients with unstable angina: association with systemic inflammation. Eur Heart J., 25:1003–1008.

George, J., Goldstein, E., Abashidze, S., Wexler, D., Hamed, S., Shmilovich, H., et al. (2005). Erythropoietin promotes endothelial progenitor cell proliferative and adhesive properties in a PI 3-kinase-dependent manner. Cardiovasc Res 68(2), 299-306.

George J, Shmilovich H, Deutsch V, Miller H, Keren G, Roth A. (2006). Comparative Analysis of Methods for Assessment of Circulating Endothelial Progenitor Cells, Tissue Engineering 12 (2) 331-335

Gerhard-Herman M, et al. (2002). Assessment of endothelial function (nitric oxide) at the tip of a finger. Circulation, 106:Suppl II:170.

Gill M, Dias S, Hattori K, Rivera ML, Hicklin D, Witte L, Girardi L, Yurt R, Himel H, Rafii S, (2001). Vascular trauma induces rapid but transient mobilization of VEGFR2/AC133 endothelial precursor cells. Circ Res., 88:167–174.

Goon, P.K.Y. Lip G.Y.H, Boos, CJ, Stonelake, PS, Blann, AD. (2006). Circulating Endothelial Cells, Endothelial Progenitor Cells, and Endothelial Microparticles in Cancer, Neoplasia, 8:79-88.

Griese DP, Ehsan A, Melo LG, Kong D, Zhang L, Mann MJ, Pratt RE, Mulligan RC, Dzau VJ, (2003). Isolation and transplantation of autologous circulating endothelial cells into denuded vessels and prosthetic grafts: implications for cell-based vascular therapy. Circulation, 108: 2710–2715.

Heeschen C, Aicher A, Lehmann R, Fichtlscherer S, Vasa M, Urbich C, Mildner-Rihm C, Martin H, Zeiher AM, Dimmeler, (2003). Erythropoietin is a potent physiological stimulus for endothelial progenitor cell mobilization. Blood, 102:1340 –1346.

Heeschen C, Lehman R, Honold J, Assmus B, Aicher A, Walter DH, Martin H, Zeiher AM, Dimmeler S. (2004). Profoundly reduced neovascularization capacity of bone marrow mononuclear cells derived from patients with chronic ischemic heart disease.Circulation, 109:1615–1622.

Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR, Crystal RG, Besmer P, Lyden D, Moore MA, Werb Z, Rafii S., (2002). Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell;109: 625-637.

Hiasa K, Ishibashi M, Ohtani K, Inoue S, Zhao Q, Kitamoto S, Sata M, Ichiki T, Takeshita A, Egashira K. Gene transfer of stromal cell-derived factor 1 enhances ischemic vasculogenesis and angiogenesis via vascular endothelial growth factor/endothelial nitric oxide synthaserelated pathway: next generation chemokine therapy for therapeutic neovascularization. Circulation, 109:2454 –2461.

Hill JM, Zalos G, Halcox JPG, Schenke WH, Waclawin MA, Quyyumi A, Finkel T. (2003). Circulating endothelial progenitor cells, vascular function and cardiovascular risk. N Engl J Med., 348:593– 600.

Hillebrands J-L, Klatter FA, van DijK WD, Rozing J. (2003). Bone marrow does not contribute substantially to endothelial-cell replacement in transplant arteriosclerosis.Nat Med., 8:194 –195.

Hu Y, Davison F, Zhan Z, Xu Q. (2003). Endothelial replacement and angiogenesis in arteriosclerotic lesions of allografts are contributed by circulating progenitor cells.Circulation, 108:3122–3127.

Hur, J., Yoon, C.H., Kim, H.S., Choi, J.H., Kang, H.J., Hwang, K.K., et al. (2004). Characterization of two types of endothelial progenitor cells and their different contributions to neovasculogenesis. Arterioscler Thromb Vasc Biol 24(2), 288–293.

Imanishi, T., Hano, T. & Nishio, I. (2005) Estrogen reduces endothelial progenitor cell senescence through augmentation of telomerase activity. J Hypertens 23(9):1699-1706.

Iwaguro H, Yamaguchi J, Kalka C, Murasawa S, Masuda H, Hayashi S, Silver M, Li T, Isner JM, Asahara T, (2002). Endothelial progenitor cell vascular endothelial growth factor gene transfer for vascular regeneration. Circulation, 105:732–738.

Kalka C, Tehrani H, Laudernberg B, Vale P, Isner JM, Asahara T, Symes JF, (2000a). Mobilization of endothelial progenitor cells following gene therapy with VEGF165 in patients with inoperable coronary disease. Ann Thorac Surg., 70:829–834.

Kalka C, Masuda H, Takahashi T, Gordon R, Tepper O, Gravereaux E, Pieczek A, Iwaguro H, Hayashi S-I, Isner JM, Asahara T (2000b). Vascular endothelial growth factor165 gene transfer augments circulating endothelial progenitor cells in human subjects. Circ Res., 86:1198 –1202.

Kalka C, Masuda H, Takahashi T, Kalka-Moll WM, Silver M, Kearney M, Li T, Isner JM, Asahara T, (2000c). Transplantation of ex vivo expanded endothelial progenitor cells for therapeutic neovascularization. Proc Natl Acad Sci U S A. 97:3422–3427.

Kang H-J, Kim H-S, Zhang S-Y, Park K-W, Cho H-J, Koo B-K, Kim Y-J, Lee DS, Sohn D-W, Han K-S, Oh B-H, Lee M-M, Park Y-B, (2004). Effects of intracoronary infusion of peripheral blood stem cells mobilized with granulocyte-colony stimulating factor on left ventrricular systolic function and restenosis after coronary stenting in myocardial infarction: the MAGIC cell randomized clinical trial. Lancet, 363:751–756.

Kawamoto A, Gwon H-C, Iwaguro H, Yamaguchi J, Uchida S, Masuda H, Silver M, Ma H, Kearney M, Isner JM, Asahara T, (2001). Therapeutic potential of ex vivo expanded endothelial progenitor cells for myocardial ischemia. Circulation, 103:634–637.

Khan SS, Solomon MA, and McCoy JP Jr, (2005). Detection of circulating-  endothelial cells and endothelial progenitor cells by flow cytometry. Cytometry B Clin Cytom64:1–8.

Kiernan, T.(2006). Endothelial progenitor cells in 2006 – Where are we now? http://www.irishheart.ie/iopen24/catalog/pub/Heartwise/2006/Spring/endothelial.pdf retrieved 6/22/2006. Link not found 5/1/2013

Kim, DH, Leu, HB, Ott, HC & Taylor, DO, Bertolini, F, Mancuso, P & Kerbel, RS, Boos, CJ, Goon, PKY, Lip, GYH, (2005). Multiple comments – Correspondence to the Editor on Circulating Endothelial Progenitor Cells. NEJM, 353:24, 2613-2616

Kleinman, ME, Blei, F, Gurtner, GC, (2005). Circulating Endothelial Progenitor Cells and Vascular Anomalies, Lymphatic Research and Biology, 3;4: 234-239.

Kocher AA, Schuster MD, Szabolcs MJ, Burkhoff D, Wang J, Homma S, Edwards NM, Itescu S. (2001). Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med., 7:430–436.

Kong D, Melo LG, Gnecchi M, Zhang L, Mostoslavski G, Liew CC, Pratt RE, Dzau VJ. (2004a). Cytokine-induced mobilization of circulating endothelial progenitor cells enhances repair of injured arteries. Circulation, 110:2039 –2046.

Kong D, Melo LG, Mangi AA, Zhang L, Lopez-Ilasaca M, Perrella MA, Liew CC, Pratt RE, Dzau VJ, (2004b). Enhanced inhibition of neointimal hyperplasia by genetically engineered endothelial progenitor cells. Circulation, 109:1769 –1775.

Kuvin JT, et al. (2003a). Assessment of peripheral vascular endothelial function with finger arterial pulse wave amplitude. Am Heart J, 146:168-74.

Kuvin JT, et al. (2003b). Peripheral arterial tonometry during hyperemia is blunted in patients with coronary artery disease. J Am Coll of Cardiology, 41:Suppl:269A.

Lapidot T, and Petit, I (2002) Current understanding of stem cell mobilization: The roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines, and stromal cells. Experimental Hematology, 30:973–98

Laufs U, Werner N, Link A, Endres M, Wassmann S, Jurgens K, Miche E, Bohm M, and Nickenig G (2004). Physical training increases endothelial progenitor cells, inhibits neointima formation, and enhances angiogenesis. Circulation 109: 220–226.

Llevadot J, Murasawa S, Kureishi Y, Uchida S, Masuda H, Kawamoto A, Walsh K, Isner JM, Asahara T, (2001). HMG-CoA reductase inhibitor mobilizes bone marrow-derived endothelial progenitor cells. J Clin Invest., 108:399–405.

Lloyd-Jones, D. and Tian, L. (2006). Predicting Cardiovascular Risk, So What Do We Do Now?. Arch Intern. Med, 166, 1342-1343.

Loomans CJM, de Koening EJP, Staal FJT, Rookmaaker MB, Verseyden C, de Boer HC, Verhaar MC, Braam B, Rebelink TJ, van Zonneveld A-J. (2004). Endothelial progenitor cell dysfunction. A novel concept in the pathogenesis of vascular complications of type I diabetes. Diabetes, 53:195–199.

Losordo DW, Isner JM, Diaz-Sandoval LJ, (2003). Endothelial Recovery. The next target in restenosis prevention. Circulation, 107:2635–2637.

Lusis, A.J. (2000). Atherosclerosis. Nature 407(6801), 233–241. DIGITAL LINK N/A

Massa M, Rosti V, Ferrario M, Campanelli R, Ramajoli, Rosso R, De Ferrari GM, Ferlini M, Goffredo L, Bertoletti A, Klersy C, Pecci A, Moratti R, Tavazzi, (2005). Increased circulating hematopoietic and endothelial progenitor cells in the early phase of acute myocardial infarction. Blood,105:199 –206.

Orlic D, Kajstura J, Chimenti S, Limana F, Jakoniuk I, Quaini F, Nadal-Ginard B, Bodine DM, Leri A, Anversa P. (2001). Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc Natl Acad Sci U S A. 98:10344 –10349.

Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, Williams M, Oz MC, Hicklin DJ, Witte L, Moore MA, and Rafii S (2000). Expression of VEGFR-2 and AC133 by circulating human CD34+ cells identifies a population of functional endothelial precursors. Blood 95: 952–958.

Perin EC, Dohmann HFR, Borojevic R, Silva SA, Sousa AL, Mesquita CT, Rossi MI, Carvalho AC, Dutra HS, Dohmann HJ, Silva GV, Belem L, Vivacqua R, Rangel FO, Esporcatte R, Geng YJ, Vaughn WK, Assad JA, Mesquita ET, Willerson JT, (2003). Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation, 107:2294 –2302.

Powell TM, Paul JD, Hill JM, Thompson M, Benjamin M, Rodrigo M, McCoy JP, Read EJ, Khuu HM, (2005). Leitman SF, Finkel T, Cannon RO III. Granulocyte colony stimulating factor mobilizes functional endothelial progenitor cells in patients with coronary artery disease. Arterioscler Thromb Vasc Biol., 25:1– 6.

Rafii S, Lyden D (2003). Therapeutic stem and progenitor cell transplantation for organ vascularisation and regeneration. Nat Med 9: 702–712.

Rauscher FM, Goldschmidt-Clermont PJ, Davis BH, Wang T, Gregg D, Ramaswami P, Pippen AM, Annex BH, Dong C, Taylor DA, (2003). Aging, progenitor cell exhaustion, and atherosclerosis. Circulation, 108:457–463.

Ross R. (1999). Atherosclerosis – An inflammatory disease. N Engl J Med., 340:115–126.

Rubanyi GM. (1993). The role of endothelium in cardiovascular homeostasis and diseases. J Cardiovasc Pharmacol., 22(Suppl):S1–S4.

Sata M, Saiura A, Kunisato A, Tojo A, Okada S, Tokuhisa T, Hirai H, Makuuchi M, Hirata Y, Nagai R. (2002). Hematopoietic stem cells differentiate into vascular cells that participate in the pathogenesis of atherosclerosis. Nat Med., 8:403– 409.

Schachinger V, Assmus B, Britten MB, Honold J, Lehman R, Teupe C, Abolmaali ND, Vogt TJ, Hofmann WK, Martin H, Dimmeler S, Zeiher AM, (2004). Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction: final one-year results of the TOPCARE-AMI trial. J Am Coll Cardiol., 44:1690 –1699.

Schatteman GC, Hanlon HD, Jiao C, Dodds SG, Christy BA. (2000). Blood derived angioblasts accelerate blood flow restoration in diabetic mice. J Clin Invest., 106:571–578.

Scheubel RJ, Zorn H, Rolf-Edgar S, Kuss O, Morawietz, Holtz J, Simm A. (2003). Age-dependent depression in circulating endothelial progenitor cells in patients undergoing coronary artery bypass grafting. J Am Col Cardiol., 42:2073–2080.

Segal, M.S., Shah, R., Afzal, A., Perrault, C.M., Chang, K., Schuler, A., et al. (2006). Nitric oxide cytoskeletal-induced alterations reverse the endothelial progenitor cell migratory defect associated with diabetes. Diabetes 55(1), 102-109

Shi Q, Raffi, Wu MH, Wijelath ES, Yu C, Ishida A, Fujita Y, Kothari S, Mohle R, Sauvage LR, Moore MAS, Storb RF, Hammond WP. (1998). Evidence of circulating bone-marrow derived endothelial cells. Blood, 92:362–367.

Shi Q, Bhattacharya V, Hong-De Wu M, Sauvage LR. (2002). Utilizing granulocyte colony-stimulating factor to enhance vascular graft endothelialization from circulating blood cells. Ann Vasc Surg., 16:314 –320.

Shintani S, Murohara T, Ikeda H, Ueni T, Honma T, Katoh A, Sasaki K, Shimada T, Oike Y, Imaizumi T, (2001). Mobilization of endothelial progenitor cells in patients with acute myocardial infarction. Circulation, 103:2776 –2779.

Shirota, T., Yasui, H., Shimokawa, H. & Matsuda, T. (2003). Fabrication of endothelial progenitor cell (EPC)-seeded intravascular stent devices and in vitro endothelialization on hybrid vascular tissue. Biomaterials 24(13), 2295–2302.

Simper D, Wang S, Deb A, Holmes D, McGregor C, Frantz R, Kushawa SS, Caplice NM, (2003). Endothelial progenitor cells are decreased in blood of cardiac allograft patients with vasculopathy and endothelial cells of non cardiac origin are enriched in transplant atherosclerosis. Circulation, 107:143–149.

Spyridopoulos I, Haendeler J, Urbich C, Brummendorf TH, Oh H, Schneider MD, Zeiher AM, Dimmeler S, (2004). Statins enhance migratory capacity by upregulation of the telomere repeat-binding factor TRF2 in endothelial progenitor cells. Circulation, 110:3136 –3142.

Stamm C, Westphal B, Kleine H-D, Petzsch M, Kittner C, Klinge H, Schumichen C, Nienaber CA, Freund M, Steinhoff G, (2003). Autologous bonemarrow transplantation for myocardial regeneration. Lancet, 361:45–46.

Strauer BE, Brehm M, Zeus T, Kostering M, Hernandez A, Sorg RV, Kogler G, Wernet P, (2002). Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation, 106:1913–1918.

Strehlow K, Werner N, Berweiler J, Link A, Dirnagl U, Priller J, Laufs K, Ghaeni L, Milosevic M, Bohm M, Nickenig G, (2003). Estrogen increases bone-marrow derived endothelial progenitor cell production and diminishes neointima formation.Circulation, 107:3059 –3065.

Takahashi T, Kalka C, Masuda H, Chen D, Silver M, Kearney M, Magner M, Isner JM, Asahara T, (1999). Ischemia- and cytokine-induced mobilization of bone-marrow-derived endothelial progenitor cells for neovascularization. Nat Med., 5:434–438.

Tepper OM, Galiano RD, Capla JM, Kalka C, Gagne PJ, Jacobwotiz GR, Levine JP, Gurtner GC. (2002). Human endothelial progenitor cells from type II diabetes exhibit impaired proliferation, adhesion, and incorporation into vascular structures.Circulation, 106:2781–2786.

Tse HF, Kwong YL, Chan JK, Lo G, Ho CL, and Lau CP (2003). Angiogenesis in ischaemic myocardium by intramyocardial autologous bone genesis marrow mononuclear cell implantation. Lancet 361: 47–49.

Tung, R, Kaul, S, Diamond, GA, Shah, PK (2006). Drug-Eluting Stents for the Management of Restenosis: A Critical Appraisal of the Evidence. Annals of Internal Medicine, 144;12: 913-919.

Valenzuela-Fernandez A, Planchenault T, Baleux F, et al. (2002) Leukocyte elastase negatively regulates stromal cell-derived factor-1 (SDF)/CXCR4 binding and functions by amino-terminal processing of SDF-1 and CXCR4.  J Biol Chem 277:156

Valgimigli M, Rigolin GM, Fucili A, Della Porta M, Soukhomovskaia O, Malagutti P, Bugli AM, Bragottu LZ, Francolini G, Mauro E, Castoldi G, Ferrari R, (2004). CD34 and endothelial progenitor cells in patients with various degrees of congestive heart failure. Circulation, 110:1209–1212.

Vasa M, Fichtlscherer S, Adler K, Aicher A, Martin H, Zeiher AM, Dimmeler S. (2001a). Increase in circulating endothelial progenitor cells by statin therapy in patients with stable coronary artery disease. Circulation, 103:2885–2890.

Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM, Dimmeler S. (2001b). Number and migratory activity of circulating endothelial progenitor cells inversely correlates with risk factors for coronary artery disease. Circ Res., 89:e1– e7.

Vasan, RS, (2006). Biomarkers of Cardiovascular Disease: Molecular Basis and Practical Considerations, Circulation, 113:2335-2362.

Verma S, Kukiszewski MA, Li S-H, Szmitko PE, Zucco L, Wang C-H, Badiwala MV, Mickle DAG, Weisel RD, Fedak PWM, Stewart DJ, Kutrik MJB, (2004). C-reactive protein attenuates endothelial progenitor cell survival, differentiation, and function.Circulation, 109:r91–r100.

Verma, S. and Marsden, P.A. (2005). Nitric Oxide-Eluting Polyurethanes – Vascular Grafts of the Future? New England Journal Medicine, 353 (7), 730-731.

Verma S, Szmitko, PE, (2006). The vascular biology of peroxisome proliferator-activated receptors: Modulation of atherosclerosis. Can J Cardiol, 22 (Suppl B):12B-17B.

Walter DH, Rittig K, Bahlmann FH, Kirchmair R, Silver M, Murayama T, Nishimura H, Losordo DW, Asahara T, Isner JM, (2002). Statin therapy accelerates reendothelialization: a novel effect involving mobilization and incorporation of bone marrow-derived endothelial progenitor cells. Circulation, 105:3017–3024.

Werner N, Junk S, Laufs L, Link A, Walenta K, Bohm M, Nickenig G., (2003).  Intravenous transfusion of endothelial progenitor cells reduces neointima formation after vascular injury. Circ Res., 93:e17– e24.

Werner N, Kosiol S, Schiegl T, Ahlers P, Walenta K, Link A, Böhm M, Nickenig G. (2005a). Circulating Endothelial Progenitor Cells and Cardiovascular Outcomes,NEJM, 353: 999-1007

Werner, N & Nickenig, G. (2005b). Authors Reply to Correspondence to the Editor on Circulating Endothelial Progenitor Cells. NEJM, 353:24, 2613-2616

Widlansky ME, et al. (2003). The clinical implications of endothelial dysfunction. J Am Coll of Cardiology, 42:1149-60.

Wollert KC, Meyer GP, Latz J, Ringes-Lichtenberg S, Lippolt P, Breindenbach C, Fichtner S, Korte T, Hornig B, Messinger D, Arseniev L, Hartenstein B, Ganser A, Drexler H (2004). Intracoronary autologous bonemarrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial. Lancet, 364:141–148.

Yamaguchi J, Kusano KF, Masuo O, Kawamoto A, Silver M, Murasawa S, Bosch-Marce M, Masuda H, Losordo DW, Isner JM, Asahara T. (2003).  Stromal cell-derived factor-1 effects on ex vivo expanded endothelial progenitor cell recruitment for ischemic neovascularization. Circulation, 107:1322–1328.

Yoon YS, Park JS, Tkebuchava T, Luedeman C, Losordo DW. (2004). Unexpected severe calcification after transplantation of bone marrow cells in acute myocardial infarction. Circulation, 109:3154 –3157.


 

Read Full Post »

Reporter: Aviral Vatsa MBBS PHD

Abstract

Wnt signaling is essential for osteogenesis and also functions as an adipogenic switch, but it is not known if interrupting wnt signaling via knockout of β‐catenin from osteoblasts would cause bone marrow adiposity. In this study the authors determined whether postnatal deletion of β‐catenin in preosteoblasts, through conditional cre expression driven by the osterix promoter, causes bone marrow adiposity. Postnatal disruption of β‐catenin in the preosteoblasts led to extensive bone marrow adiposity and low bone mass in adult mice. In cultured bone marrow‐derived cells isolated from the knockout mice, adipogenic differentiation was dramatically increased, whereas osteogenic differentiation was significantly decreased. As myoblasts, in the absence of wnt/β‐catenin signaling, can be reprogrammed into the adipocyte lineage, we sought to determine whether the increased adipogenesis we observed partly resulted from a cell‐fate shift of preosteoblasts that had to express osterix, (lineage‐committed early osteoblasts), from the osteoblastic to the adipocyte lineage. Using lineage tracing both in vivo and in vitro we demonstrated that the loss of β‐catenin from preosteoblasts caused a cell‐fate shift of these cells from osteoblasts to adipocytes, a shift that may at least partly contribute to the bone marrow adiposity and low bone mass in the knockout mice. These novel findings indicate that wnt/β‐catenin signaling exerts control over the fate of lineage‐committed early osteoblasts, with respect to their differentiation into osteoblastic vs. adipocytic populations in bone, and thus offers potential insight into the origin of bone marrow adiposity. © 2012 American Society for Bone and Mineral Research.

Read Full Post »

%d bloggers like this: