JY Rev Circ Res 2010

June 14, 2017 | Autor: Yagna Jarajapu | Categoria: Pharmacology, Pharmacy
Share Embed


Descrição do Produto

The Promise of Cell-Based Therapies for Diabetic Complications: Challenges and Solutions Yagna P.R. Jarajapu and Maria B. Grant Circ. Res. 2010;106;854-869 DOI: 10.1161/CIRCRESAHA.109.213140 Circulation Research is published by the American Heart Association. 7272 Greenville Avenue, Dallas, TX 72514 Copyright © 2010 American Heart Association. All rights reserved. Print ISSN: 0009-7330. Online ISSN: 1524-4571

The online version of this article, along with updated information and services, is located on the World Wide Web at: http://circres.ahajournals.org/cgi/content/full/106/5/854

Subscriptions: Information about subscribing to Circulation Research is online at http://circres.ahajournals.org/subscriptions/ Permissions: Permissions & Rights Desk, Lippincott Williams & Wilkins, a division of Wolters Kluwer Health, 351 West Camden Street, Baltimore, MD 21202-2436. Phone: 410-528-4050. Fax: 410-528-8550. E-mail: [email protected] Reprints: Information about reprints can be found online at http://www.lww.com/reprints

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

This article is part of a thematic series on Cardiovascular Complications of Diabetes and Obesity which includes the following articles: The Impact of Macrophage Insulin Resistance on Advanced Atherosclerotic Plaque Progression [2010;106:58 – 67] The RAGE Axis: A Fundamental Mechanism Signaling Danger to the Vulnerable Vasculature [2010;106:842– 853] The Promise of Cell-Based Therapies for Diabetic Complications: Challenges and Solutions

Oxidative Stress and Diabetic Complications The Polyol Pathway: Implications for Atherosclerosis and Cardiac Dysfunction in Diabetes Protein Kinase C Isoforms & the Impact of Diabetic Complications ER Stress, Inflammation, Obesity, and Diabetes Epigenetics - Mechanisms and Implications for Diabetic Complications

Ann Marie Schmidt, Guest Editor

The Promise of Cell-Based Therapies for Diabetic Complications Challenges and Solutions Yagna P.R. Jarajapu, Maria B. Grant Abstract: The discovery of endothelial progenitor cells (EPCs) in human peripheral blood advanced the field of cell-based therapeutics for many pathological conditions. Despite the lack of agreement about the existence and characteristics of EPCs, autologous EPC populations represent a novel treatment option for complications requiring therapeutic revascularization and vascular repair. Patients with diabetic complications represent a population of patients that may benefit from cellular therapy yet their broadly dysfunctional cells may limit the feasibility of this approach. Diabetic EPCs have decreased migratory prowess and reduced proliferative capacity and an altered cytokine/growth factor secretory profile that can accelerate deleterious repair mechanisms rather than support proper vascular repair. Furthermore, the diabetic environment poses additional challenges for the autologous transplantation of cells. The present review is focused on correcting diabetic EPC dysfunction and the challenges involved in the application of cell-based therapies for treatment of diabetic vascular complications. In addition, ex vivo and in vivo functional manipulation(s) of EPCs to overcome these hurdles are discussed. (Circ Res. 2010;106:854-869.) Key Words: diabetes 䡲 endothelial progenitor cells 䡲 bone marrow 䡲 angiogenesis

D

iabetes is associated with a broad spectrum of vascular complications that constitute a major health care concern in the western world. Despite the prevalence and the debilitating nature of diabetic vascular complications, cellular/molecular/genetic mechanisms underlying vascular dysfunction remain unclear and vary with the cell type studied and the vascular bed examined; however, the literature to date reflects a consensus that endothelial dysfunction is a key initiator that precedes development of vascular complications. Almost 6000 research articles have been published

since 1973 that directly link diabetic vascular complications to endothelial dysfunction. These publications report studies using animal models as well as those describing clinical trials and their outcomes. The definition of endothelial dysfunction varies depending on the organ studied; however, in general, endothelial dysfunction can be characterized by impaired endotheliumdependent dilatation to agonists, to shear stress, or to local ischemia. In a particular context such as proliferative diabetic retinopathy (PDR), the dysfunction is described as endothe-

Original received November 12, 2009; revision received January 29, 2010; accepted February 1, 2010. From Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville. Correspondence to Maria B. Grant, MD, Pharmacology and Therapeutics, College of Medicine, University of Florida, PO Box 100267, Gainesville, FL 32610-0267. E-mail [email protected] © 2010 American Heart Association, Inc. Circulation Research is available at http://circres.ahajournals.org

DOI: 10.1161/CIRCRESAHA.109.213140

854 at Univ Florida on March 19, 2010 Downloaded from circres.ahajournals.org

Jarajapu and Grant lial activation with proinflammatory and proliferative phenotype that ultimately disrupts vascular integrity resulting in increased permeability. In general, diabetic microvascular complications are typically associated with dysregulation of vascular remodeling and vascular growth with decreased responsiveness to ischemic/hypoxic stimuli and impaired or abnormal neovascularization. Lack of endothelial regeneration and impaired angiogenesis contribute to the progression of diabetic micro and macrovascular complications. Formation of stable vasculature in response to tissue injury is an essential event for the restoration of blood flow and the repair of the affected tissue areas. Currently, clinical management of diabetic complications relies exclusively on pharmacological therapeutics that minimally affect endothelial repair or regeneration in most cases, and, therefore, these treatments have modest influence on end organ dysfunction. Hence, there is a need for therapeutic interventions that actually accelerate repair of dysfunctional endothelium in the end organ and restore blood flow to result in functional tissue generation. After the identification of “putative” endothelial progenitor cells (EPCs),1 diabetic vascular complications are now thought to be, in part, the result of the reparative dysfunction of these cells. In response to these intriguing findings, a rapid progression of EPCs from the “bench to the bedside” occurred via translational studies even in the absence of a consensus about the true identity of the “EPC.” These seminal discoveries indeed paved the way for cell-based therapeutics for patients with vascular complications; however, a few caveats must be considered for autologous transplantation of cells in diabetic patients. The cells themselves are known to be dysfunctional, and the diabetic environment poses a variety of challenges that need to be considered. Otherwise, cell-based therapies may not be beneficial and may actually result in worsening of existing vascular complications in a patient population most in need of help. The present review provides a brief introduction to the characteristics of EPCs and their dysfunction in diabetes and focuses mainly on the challenges involved in the application of cell-based therapies for diabetic vascular complications. In addition, a variety of possible ex vivo and in vivo manipulation(s) of EPCs to overcome these hurdles are discussed.

Putative EPCs Based on the observations that Dacron arterial prosthesis in humans were endothelialized,2,3 it was hypothesized that a subpopulation of cells in the circulation support the replenishment of endothelial cells (ECs) within the vasculature and, in part, sustained neovascularization in response to tissue injury. This concept was similar to the phenomenon of hematopoietic recovery and persistent replenishment of blood cells by circulating hematopoietic stem cells (HSCs). Before these observations, adult neovascularization or de novo formation of blood vessels was thought to occur exclusively by angiogenesis, which is formation of blood vessels by existing vessels, rather than by vasculogenesis, formation of blood vessels from stem or progenitor cells (Figure 1).4,5 This long-held paradigm regarding the origins of neovascularization was shifted by the observations of Asahara et al1

Cell-Based Therapies for Diabetic Complications

855

Non-standard Abbreviations and Acronyms AGE BM BMC CFU CXCR EC eEPC eNOS EPC G-CSF GM-CSF HSC IGF iNOS MAPK MMP MSC NPDR OEC PDR ROS SDF SMC STZ VEGF VEGFR

advanced glycation end products bone marrow bone marrow– derived progenitor cell colony-forming unit CXC chemokine receptor endothelial cell early endothelial progenitor cell endothelial NO synthase endothelial progenitor cell granulocyte colony-stimulating factor granulocyte/macrophage colony-stimulating factor hematopoietic stem cell insulin-like growth factor inducible NO synthase mitogen-activated protein kinase matrix metalloproteinase mesenchymal stem cell nonproliferative diabetic neuropathy outgrowth endothelial cells proliferative diabetic neuropathy reactive oxygen species stromal cell– derived factor smooth muscle cell streptozotocin vascular endothelial growth factor vascular endothelial growth factor receptor

This study showed that the putative EC progenitor cells or angioblasts could be isolated from human peripheral blood by magnetic bead selection on the basis of cell surface antigen expression and in vitro these cells differentiated into ECs. Specifically, CD34⫹VEGFR-2⫹ mononuclear cells were shown to express EC-associated markers such as CD31, vascular endothelial growth factor (VEGF) receptor (VEGFR)-2, Tie-2, and E-selectin when cultured on fibronectin. The EPC population of Asahara et al1 also expressed endothelial NO synthase (eNOS). However, both CD34 and VEGFR-2 antigens are present on ECs, and so, to exclude differentiated circulating ECs from a study population, now CD133 has been included as a marker limited to primitive cells but not expressed on ECs. To date, many antigenic markers including CD34, CD133, CD45, VEGFR-2, CD133, CXC chemokine receptor (CXCR)4, CD14, and CD31 have been used to identify EPC populations. Cell populations characterized by these surface markers have been shown to become angiogenic or to support angiogenesis in in vitro or in vivo assays.6 –11 An elegant study by Loomans et al12 in mice suggested eNOS expression is a reliable marker to identify bone marrow (BM)-derived EPCs and this concept has been strongly supported by other studies.1,13 In animal models of ischemia, heterologous, homologous, and autologous EC progenitors incorporated into sites of active angiogenesis, and this led to their use for augmentation of collateral vessel

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

856

Circulation Research

March 19, 2010

Figure 1. Adult stem cells of the bone marrow: the BM hosts at least 2 known types of adult stem cells, the HSCs and the MSCs. The MSCs have been shown to differentiate into various cell types, including osteoblasts, adipocytes, chondrocytes, myocytes, fibroblasts and endothelial cells. The most prominent adult stem cell in the BM is the HSC. HSCs can give rise to the hematopoietic progenitor cells, which in turn give rise to the lymphoid, the myeloid and likely the EPC. The BM microenvironment is composed of BM stromal cells (which are the source of SDF-1), adipocytes, and cells of the bone matrix, osteoblasts, and osteoclasts. The vessels within the BM, composed of pericytes and endothelium, function to provide a barrier between the hematopoietic compartment and the circulatory system discussed in greater detail in Figure 2.

growth to ischemic tissues. These pivotal studies were corroborated by many investigators and by the discovery that these EPCs can be found in BM niches and mobilized to areas of tissue injury/repair (Figure 2).14,15 Later, the protocol of Asahara et al was slightly modified by eliminating the cell-sorting step and the so-called colony-forming unit (CFU)-Hill or CFU-EC was developed as a commercial kit to quantify EPCs and enumeration of CFU-EC correlated closely with cardiovascular risk factors.16 Recent studies examining the relationship between EPC number/ function and presence of vascular disease support that CD34⫹ cells or cells that express both CD34 and VEGFR-2 are reliable markers for the “putative” EPC phenotype and that cells with these surface markers indeed predict defective vasoregenerative capacity in a given clinical condition. CD34⫹VEGFR-2 cell counts predicted the occurrence of cardiovascular events in a 10 month follow-up study involving 120 patients.17 CD133⫹ cell counts also predicted cardiovascular events, but to a lesser degree in a study involving 519 patients with 12month follow-up.18 Patients with coronary artery disease showed a 5-fold reduction in three different subsets of circulating CD133⫹ cells: CD133⫹CD34⫹, CD133⫹CXCR4⫹, and CD133⫹VEGFR-2⫹ cells. Total CD34⫹ cells correlated with all cardiovascular parameters and risk estimates better than CD34⫹VEGFR-2 and CD133⫹-based phenotypes.19,20 Recently, Fadini et al21 reported that circulating CD34⫹ cell number was an independent risk biomarker of cardiovascular events and significantly correlated with outcomes in metabolic syndrome, based on their study involving 214 patients over a 34 month period. Taken together, CD34⫹ cells clearly

represent a cell population of clinical utility. Although CD34⫹VEGFR-2 ⫹ represent an equally efficient marker, the number of these cells found in steady-state peripheral blood is extremely low. Despite the convincing clinical evidence, no studies have demonstrated that CD34⫹VEGFR-2 or CD34⫹CD133⫹VEGFR-2⫹ cells differentiate into EC cells in vitro.22 Although direct vascular integration of CD34⫹ cells has been shown in some in vivo studies, more studies attribute the reparative function of these cells not to direct integration but to paracrine mechanisms (Table).10,23–28 Recently, the assertion that CD34⫹VEGFR-2⫹ cells are bona fide EPCs has also been brought under fire.22 The cells growing as CFU-Hill colonies have been suggested to be mostly composed of monocytes and T cells29 and thus genetically linked to primitive hematopoietic cells.30 Later studies also revealed that CFU-ECs are not endothelial-committed and do not form perfused vessels in vivo.30 Monocytes have recently been shown to acquire endothelial markers in vitro by the uptake of platelet microparticles and thus a simple transfer of CD31 and vWF antigens can occur.31 Alternatively EPC populations can be isolated from peripheral or umbilical cord blood using in vitro culture producing 2 distinct subtypes which have been named early EPCs (eEPCs) and outgrowth ECs (OECs).32 This ex vivo analysis of cells by expansion culture has become popular and is now widely used for characterizing EPCs. These 2 phenotypically different populations are isolated from total mononuclear cells based on the time in culture and the matrices used: eEPCs originate as early as 4 days after plating and organize into clusters with very low proliferative potential and OEC (or late outgrowth EPCs) survive up to 2 to 4 weeks and exhibit endothelial morphology with a higher proliferative potential.32,33 eEPCs are a heterogeneous population of cells and are believed to be hematopoietic in origin because these cells display overlapping markers for ECs, monocytes, or macrophages; are phagocytotic and antithrombogenic; and give rise to macrophages. These cells typically do not participate in vascular repair but have the potential to augment vascular network formation by secreting paracrine factors. OECs express endothelial markers, lack CD14 or CD45, form tubular structures de novo, and incorporate into developing vascular networks but are believed to be devoid of paracrine effects.30,32,34,35 EPCs analyzed by flow cytometry and those obtained from in vitro expansion cultures have phenotypic/functional characteristics in common; however, these characteristics have not been thoroughly compared. Schofield36 suggested that “stem cell” properties can be imposed on cells that normally do not possess stem cell function by an “appropriate” microenvironment and a corollary we put forth is that an “inappropriate” microenvironment can result in a loss of a cell’s stem cell/progenitor properties. In health, a range of cells may have this stem cell potential to some extent. In contrast to freshly isolated cells, EPCs and circulating angiogenic cell colonies derived from blood samples were higher in number in coronary artery disease patients compared to controls, and the number of colonies increased with the severity of disease, instead of decreasing, as might be expected on the basis of studies in freshly isolated CD34⫹ cells.37,38 Similarly, no difference was observed in the in vitro

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

Jarajapu and Grant

Cell-Based Therapies for Diabetic Complications

857

Figure 2. BM and HSC niches: the maintenance of HSC self-renewal and differentiation is dependent on the specialized microenvironments or “niches.” Stem cells are known to reside in close proximity to endosteal linings of the BM cavities, endosteal niche, or close to sinusoidal endothelium, the vascular niche. Blood capillaries or BM sinusoids drain into a central sinus, the largest vascular structure in the BM, which contains more committed stem and progenitor cells than the relatively quiescent stem cells of the endosteal niche. Mobilization of BM cells involves the exodus of stem/progenitor cells into the circulation, whereas homing is the “opposite” of this event. HSCs mobilize from the endosteal niche move to the vascular niche and ultimately into the circulation. Mobilization is dependent on levels of cytokines or growth factors such as SDF-1, G-CSF, fibroblast growth factor (FGF), or VEGF in the BM and circulation with the involvement of matrix metalloproteinases such as MMP-2, MMP-9, cathepsin-G, and elastase. Homing involves interaction of integrins on HSCs/EPCs that are stimulated by SDF-1 with vascular cell adhesion molecule (Vcam), intercellular adhesion molecule, E-, and P-selectins expressed on BM endothelial cells (BMEC), followed by firm adhesion and subsequent endothelial transmigration into the hematopoietic compartment is mainly accomplished by very-late antigen-4 (VLA-4) interactions. HPC indicates hematopoietic progenitor cells; MPP, multipotent progenitor cells; mSCF, murine stem cell factor; Opn, osteopontin; SNO, S-nitrosothiol. Copyright 2007 American Diabetes Association. From Diabetes 2007;56:960 –967. Reprinted with permission from The American Diabetes Association.

endothelial differentiation from BM-derived cells from normal and 3 month streptozotocin (STZ)-diabetic rats.39 These studies raise questions about the value of culture-expanded EPCs as valid diagnostic or prognostic tools. Despite the lack of consensus on the identity of EPCs based on surface expression of antigenic markers and the clinical relevance of in vitro expanded progenitor populations, most investigators agree that the following criteria must be satisfied by a cell in order for it to be considered an EPC. The cell must mobilize from niches in response to ischemic stimuli, homing to areas of ischemia and participation in neovessel formation. Although there is an ongoing debate about the exact identity of EPCs, evidence has been accumulating from preclinical and clinical studies that multiple populations of progenitor cells whether freshly isolated or in vitro expanded may have therapeutic efficacy (Table).40 – 43

Paracrine Function of EPCs The degree of vascular engraftment by administered putative EPCs/BM-derived progenitor cells (BMCs) varies among studies but ranges from none to a high percentage.1,13,25,26 Regardless of the number of cells that physically integrate

into the vessel wall and the antigenic/phenotypic characteristics they express, circulating BMCs participate in vascular repair and promote vascular growth by releasing proangiogenic factors. An elegant study by Majka et al44 reported that human CD34 ⫹ cells, myeloblasts, erythroblasts, and megakaryoblasts release numerous growth factors, cytokines, and chemokines that regulate the process of hematopoiesis by autocrine and paracrine mechanisms; however, this study did not evaluate vascular repair. Harraz et al13 showed that CD34⫺CD14⫹ or CD34⫺ cells can incorporate into the endothelium of blood vessels in mouse ischemic limbs; however, this required the presence/coinjection of CD34⫹ cells, clearly indicating paracrine modulation of CD14⫹ populations by CD34⫹ populations. Direct evidence for the paracrine interaction between subsets of EPCs was also provided by Krenning et al,45 who showed that CD34⫹ cells modulate proliferation and endothelial differentiation of CD14 ⫹ cells by releasing hepatocyte growth factor, interleukin-8, and monocyte chemoattractant protein-1. In yet another study, culture-expanded EPCs derived from human monocytes/macrophages exhibited low potential for proliferation and endothelial differentiation but were capable of

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

858 Table.

Circulation Research

March 19, 2010

Antigenic Characteristics of Different Progenitor Populations That Have Been Evaluated for Therapeutic Angiogenesis

Antigenic Markers

Donor

Test System(s)

Functional End Point(s)

Proposed Mechanism(s)

Mouse or Rabbit HLI

None

Direct Differentiation1

Diabetic mouse HLI

Limb perfusion,

Vascular incorporation26

Rat MI

Capillary density, myocardial function and remodeling

Direct differentiation and paracrine effects161

CD34lowcKIT⫹Sca1⫹

Mouse MI

None

Direct differentiation162

CD34⫺ or CD34⫺CD14⫹

Mouse HLI

Limb perfusion

Direct differentiation and vascular incorporation13

Diabetic mouse wounds

Vascularity

Paracrine effects23

Mouse MI

Infarct size, myocardial perfusion

Direct differentiation163

Mouse HLI

Limb perfusion and autoamputation54

Diabetic ischemic wounds in mice

Wound closure

Paracrine effects48

Diabetic mouse HLI

Limb perfusion

Paracrine effects10

Mouse MI

Myocardial function

Direct differentiation and vascular incorporation164

Rabbit carotid balloon injury

Endothelial function; neointima formation

Direct differentiation165

Mouse HLI

Limb perfusion

Paracrine effects; direct differentiation or vascular incorporation166–169

CD34⫹/CD45⫹/Flk-1⫹ CD34⫹

Human Diabetic

CD34⫹CD117⫹GATA⫹

CD34⫹ or CD34⫺ CD34⫹ or CD34⫹KDR⫹ CD34⫹ CD133⫹ CD34⫹ or CD14⫹ Culture-expanded cells

Obese-diabetic mouse

Refer to the recent review by Sekiguchi et al42 for preclinical studies in models of MI and controlled clinical trials.

secreting proangiogenic factors VEGF, hepatocyte growth factor, granulocyte colony-stimulating factor (G-CSF), and granulocyte/macrophage (GM)-CSF.46 Conditioned medium of culture-expanded EPCs produced a strong migratory response in ECs in vitro that was abolished by neutralizing antibodies to stromal cell– derived factor (SDF)-1 and VEGF.47 Recently, CD133⫹ cells were shown to secrete interleukins, growth factors and chemokines that are capable of accelerating vascular network formation in vivo and enhancing the healing of ischemic ulcers in diabetic mice.48 For a detailed account of paracrine mechanisms in adult stem cell signaling and therapy, refer to the recent reviews by Gnecchi et al49 and Burchfield and Dimmeler.50 Studies by Katusic and colleagues provided a mechanistic basis for the paracrine modulation of vascular function and accelerated angiogenesis by EPCs. In their studies, cultureexpanded human EPCs released interleukin-8 and demonstrated a mitogenic effect on vascular ECs.51 Paracrine factors released by ex vivo expanded rabbit EPCs conferred cerebrovascular protection by increasing prostaglandin I2 production via cyclooxygenase-2/prostaglandin I2 synthase and by reducing thromboxane A2 production but did not change eNOS or inducible (i)NOS levels.52 Culture-expanded human EPCs were shown to express cyclooxygenase-1 and secrete prostaglandin I2 that increase angiogenesis via activation of peroxisome proliferator-activated receptor-␦.53 Paracrine function of EPCs in diabetes has not been extensively studied, but the literature suggests that diabetic EPCs have impaired paracrine function in vitro and in vivo

(Figure 3). Diabetic EPCs may not be able to release adequate levels of factors in the diabetic environment or alternatively, the diabetic environment may degrade these factors before they are able to promote neovascularization. Awad et al54 reported an interesting observation that obesity and diabetes together differentially affect primitive CD34⫹ and monocytic CD14⫹ cell function and furthermore, convert the proangiogenic phenotype to antiangiogenic in CD34⫹ cells, whereas CD14⫹ cells are affected to a lesser extent. These findings can be explained on the basis of paracrine release of antiangiogenic factors by cells of obese/diabetic origin. We observed that the release of SCF and hepatocyte growth factor by diabetic CD34⫹ cells in to the conditioned medium was lower whereas the secretion of transforming growth factor-␤1 was higher compared to the nondiabetic cells, and, moreover, conditioned medium of diabetic cells did not support the proliferation and migration of nondiabetic cells in vitro.55

Circulating Stem/Progenitor Cells in Diabetes Diabetic individuals with vascular complications would potentially benefit from cellular therapy with autologous cells; however, their EPCs are dysfunctional, because of a reduction in the number of circulating progenitors56 –59 or attenuated function in in vitro angiogenic assays with or without an actual reduction in the number of cells.60,61 In a series of systematic studies, Egan et al59 evaluated cellular phenotypes in the peripheral blood of diabetic patients and observed a significant decline in the number of CD34⫹, CD34⫹ KDR⫹, CD34⫹CD133⫹KDR⫹, CD133⫹KDR⫹, CD117⫹KDR⫹,

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

Jarajapu and Grant

Figure 3. Diabetic dysfunction in the BM mobilization of stem/progenitor cells and paracrine regulation of ischemic vascular repair. In normal conditions, factors released by ischemic/injured tissue causes mobilization of BM cells, which, when transmigrated into areas of ischemia, release proangiogenic factors and physiological levels of NO and ROS that modulate repair mechanisms by activating vascular endothelium in the surrounding areas, by recruiting more BM cells and by modifying ischemic environment. In diabetic conditions, signals arising from the vascular injury are weaker, resulting in reduced mobilization of BM cells into circulation. Those few progenitor cells reaching the areas of ischemia are either not able to release proangiogenic factors or release antiangiogenic or proinflammatory factors and nonphysiological levels of NO and ROS that delay or inhibit vascular repair.

and CD34⫹CD31⫹ cell populations compared to normal subjects. These results clearly indicate a generalized reduction in putative endothelial progenitors with different levels of maturity/differentiation. In contrast, Lee et al62 demonstrated an increase in c-Kit⫹ and CD34⫹ cells in advanced PDR; however, the results were based on a small sample size and a limited characterization of comorbidities and drug treatment that may have had an effect on progenitor numbers. Kusuyama et al63 showed that in newly diagnosed patients with type 1 diabetes, the number of EPCs was significantly related to hemoglobin A1c and blood sugar levels before treatment and, therefore, concluded that diabetes reduces the number of circulating EPCs according to its severity. The decline in the number of circulating progenitor cells is linearly correlated with the severity of diabetes56,59 and degree of glucose control has been shown to be negatively correlated with circulating progenitor numbers.57,63 Therefore, it is clear that improvement of glycemic control significantly increases the number of circulating EPCs which are vascular protective. Similar observations have been made in experimental studies that reported decreased numbers of BMCs in circula-

Cell-Based Therapies for Diabetic Complications

859

tion of diabetic rodents in response to ischemic injury compared to nondiabetic animals and this was associated with reduced eNOS expression in these cells.64,65 The reduction in circulating progenitor cells may signal diabetic dysfunction of BM or an inability to respond to signals from injured vasculature/ischemic tissue (Figure 3). Selective depletion of functional hematopoietic progenitor cells in the BM was also observed in the patients with postinfarction heart failure, suggesting increased demand and turnover. In contrast, Nguyen et al66 found an increased number of myofibroblast progenitors in these patients, suggesting that the reduction may be limited to cardio- and vasculoprotective progenitors. Thus, reduced numbers of functional EPCs in circulation have been identified as an important pathology in diabetic patients. Diabetic BM dysfunction has been poorly studied. Diabetic neuropathy may be a determinant of this diabetic BM defect. We recently reported that decreased numbers of BMCs in the circulation are accompanied by increased numbers of “trapped” cells in the BM of type-2 diabetic rats. In physiological conditions, the release of BMCs into the circulation follows a definitive circadian pattern, which is regulated by sympathetic neuronal activity. Diabetic peripheral neuropathy affects the BM and is manifested as a reduced number of nerve endings within the BM. This leads to an altered circadian rhythmicity of BMC release and reduced numbers of cells in the circulation.67 Changes in signaling of matrix metalloproteinase (MMP)-9, eNOS (see below), and c-kit ligand–receptor may also mediate this BM dysfunction and involvement of additional factors cannot be ruled out. The diabetic defect in the release of BM cells is reversed by glucose normalization early in the disease course, suggesting that the mobilization mechanisms are sensitive to chronic hyperglycemic conditions and early on remain reversible.58,64,68 In contrast, mobilizing cells from BM is not always an ideal option as cells obtained after G-CSF stimulation were found to have reduced in vivo vasoreparative function in patients with ischemic heart disease despite their higher clonal expansion capacity.69 Moreover, mobilization of BMCs for vasoreparation was shown to aggravate underlying vascular dysfunction in some clinical conditions.70 Mobilized cells from diabetic BM have not yet been characterized but need to be investigated systematically to identify the risk/benefit ratio.

Diabetes and EPC Dysfunction Diabetic EPCs exhibit reduced proliferative potential and migratory function.25,26,60,71 Vascularization was depressed when diabetic EPCs were injected into normal mice.25,72 Moreover, diabetic EPCs show a reduced ability to integrate into EC tubes in vitro.61 A large number of studies point to the defective NO signaling as a mediator of diabetic EPC dysfunction.

NO and Oxidative Stress NO is generated from the guanidino group of L-arginine and is an NADPH-dependent reaction catalyzed by a family of NO synthase (NOS) enzymes: eNOS, neuronal NOS, and iNOS. Deficiencies in L-arginine supply have been strongly implicated in vascular diseases, including diabetes. If the

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

860

Circulation Research

March 19, 2010

supply of L-arginine or the essential cofactor tetrahydrobiopterin is inadequate, NOS becomes “uncoupled” and uses molecular oxygen as a substrate to form superoxide instead of NO. An imbalance between L-arginine availability and NOS activity can also occur when cellular transport of L-arginine is inhibited,73 recycling of L-citrulline back to L-arginine is reduced, catabolism of L-arginine by arginase is increased,74 or competitive inhibition for the active site of NOS by asymmetrical dimethylarginine occurs.75 In diabetic vasculature, eNOS was decreased, whereas NADPH oxidase (a major source of superoxide in the vascular endothelium), iNOS, and ONOO⫺ were all increased.76 The shift in redox state in diabetes with increased generation of reactive oxygen species (ROS) causes interaction of NO with superoxide, resulting in loss of bioavailable NO and formation of ONOO⫺. The reduced NO and elevated ONOO⫺ can lead to microvascular dysfunction in diabetes.77–79 In diabetes, uncoupling of eNOS has been found to be a source of ROS generation and, when blocked, mouse models show reduced signs of endothelial dysfunction.80 Diabetic vascular dysfunction in rats was shown to be prevented by administration of 1400W, a specific iNOS inhibitor, suggesting a major role for the iNOS in diabetic complications.81,82 NO-mediated signaling pathways are essential for mobilization of BMCs83,84; NO activates MMP-9, releasing soluble Kit ligand, which shifts resident BM cells from a quiescent to a proliferative niche and stimulates rapid mobilization into the circulation.84,85 NO regulates migration of stem/progenitor cells into ischemic sites and promotes their survival.86,87 It has been shown that diabetic BMCs have decreased eNOS activity, and more importantly, that exogenous NO can correct the migratory defect in these cells.71 In addition to its effects on NO signaling, hyperglycemia has been shown to decrease circulating BMCs by increasing their senescence via activation of p38 mitogen-activated protein kinase (MAPK)88 or Akt/p53/p2189 pathways or by downregulation of SIRT1, the mammalian homolog of Sir2 (silent information regulator-2).90 A recent study showed that hyperglycemia shifts differentiation of BMCs into proinflammatory phenotype with decreased EPCs and increased macrophages.91

Increased ROS in EPC Dysfunction In physiological conditions, ROS have been shown to be involved in cellular signaling mechanisms that result from the reversible oxidation of redox-sensitive target proteins. Protein tyrosine phosphatases are extremely sensitive to oxidative modification which leads to increased phosphorylation and activation of many receptor tyrosine kinases.92 Overproduction of ROS in diabetes caused by increased activation of NADPH oxidase has been shown to be involved in the initiation and progression of diabetic vascular complications by decreasing the bioavailability of NO.93,94 BMCs express NADPH oxidase isoforms and it has been suggested that low levels of ROS in the BM play an essential role in preserving primitive HSCs in the hypoxic environment; slightly elevated levels promote mobilization of HSCs in the early stages of postischemic neovascularization, but excessive ROS production causes senescence and impairs

self-renewal of HSCs.95–98 Expression of antioxidant enzymes catalase, glutathione peroxidase, and manganese superoxide dismutase (MnSOD) is higher in circulating EPCs, whereas basal levels of ROS are lower compared to that in ECs, suggesting that EPCs are resistant to oxidative stress.99,100 In contrast, Ingram et al101 reported that clonogenic proliferative EPCs derived from adult peripheral blood are more sensitive to oxidative stress and exhibit decreased clonogenic capacity and angiogenic function in the presence of oxidants. Our studies show that human CD34⫹ cells of diabetic origin show higher levels of NADPH oxidasedependent ROS production and decreased NO bioavailability. We further observed that decreasing NADPH oxidase activation results in reduced ROS production, increased bioavailable NO levels, and improved EPC migration in response to SDF-1 or VEGF.102

Advanced Glycation End Products Advanced glycation end products (AGEs) are modified proteins or lipids that are nonenzymatically glycated and oxidized after chronic exposure to sugars associated with diabetes. AGEs affect cellular function by altering intracellular mechanisms via activating the receptor for AGEs, RAGE, as well as by modifying the extracellular environment by the formation of cross-links between key molecules in the basement of membrane/extracellular matrix. Several studies suggested that AGEs directly impair the vasoreparative function of EPCs. Incorporation of CD34⫹ cells into endothelial sprouting was impaired in the presence of pathological AGE concentrations because of enhanced apoptosis of cells via activation of p38 and p44/42 MAPKs or extracellular signalregulated kinase 1/2 MAPK pathways with the activation of nuclear factor ␬B.103,104 Other studies implicated oxidative stress and downregulation of Akt and cyclooxygenase-2 in the AGE-mediated apoptosis, impaired migration, and reduced tube formation in EPCs that were reversed by anti-RAGE antibody.105,106 In diabetes, the modification of extracellular matrix proteins by AGEs may impair or prevent interaction of EPCs with the vascular wall, thereby adversely affecting the initial triggering events in the process of re-endothelialization and angiogenesis.107 This may be the case when autologous cells are “corrected” for their diabetic dysfunction ex vivo before their reinfusion for therapy. This is one of the challenges that need to be addressed when autologous cell transplantation is considered for therapeutic reendothelialization in diabetes. For a detailed account of the biological effects of AGES and strategies to alleviate their effects in diabetes refer to the recent review by Negre-Salvayre et al.108

Diabetic Microvascular Complications Retinopathy Retinopathy is the most common of all diabetic complications, with almost all diabetic patients developing background retinopathy or nonproliferative diabetic retinopathy (NPDR), the non–vision-threatening form of diabetic retinopathy that is characterized by “microaneurysms,” hemorrhages, and exudates. The later stages of NPDR are accompanied by vasodegeneration characterized by retinal ischemia attribut-

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

Jarajapu and Grant able to areas of nonperfusion. These ischemic areas appear as acellular capillaries in the histological examination, a hallmark feature of diabetic retinopathy. As discussed in the preceding sections, diabetes is associated with both widespread dysfunction and reduced numbers of EPCs. Thus, it is not surprising that several studies have detected reduced EPC number in patients with NPDR.62,63 Brunner et al109 conducted a case– control study that compared 90 patients with type 1 diabetes with and without retinopathy and concluded that in type 1 with retinopathy, EPCs underwent stage-related regulation. In NPDR, a reduction of EPC numbers was observed, whereas in PDR, a dramatic increase of mature EPCs was observed. This is in keeping with the hypothesis that the vasodegenerative phase of NPDR is associated with reduced reparative function of these cells. In contrast, Lee et al62 demonstrated an increase in c-Kit⫹CD34⫹ cells in advanced PDR; however, the study was small in sample size and there was limited characterization of comorbidities and drug treatments that could influence progenitor numbers. Kusuyama et al examined EPCs from peripheral blood obtained from 11 patients with type 1 diabetes with PDR, 12 age- and gender-matched type 1 diabetics without retinopathy, and 11 age- and sex-matched nondiabetic controls.63 They found that the number of colonyforming units per 1⫻106 monocytes was increased in patients with PDR when compared with patients without retinopathy. Nondiabetic controls showed an intermediate number of EPC counts that were nonetheless significantly increased with respect to patients without retinopathy. Taken together, these results can be explained by the hypothesis that in contrast to EPCs in healthy individuals that rescue and maintain the existing retinal capillary bed, in diabetic patients, the reduced number and reduced clonogenic potential of EPCs might predispose these patients to development of the vasodegenerative phase of diabetic retinopathy. Once the damage is widespread, inflammation increases and chemokines are produced by the ischemic retina; the BM may respond/compensate by increasing the production of EPCs, which, in the pathological setting of a growth factor-rich vitreous and retina, may result in aberrant neovascularization characteristic of PDR. No studies to date have evaluated the potential of EPCs in the treatment of patients with diabetic retinopathy. However, animal studies have provided a better understanding of possible vascular repair by EPCs in retinopathy. Participation of BM cells in the retinal repair has been documented in mouse models; homing to areas of retinal injury and differentiation of BMCs into ECs, microglia, and astrocytes have been observed.110 –113 High numbers of EPCs contributing to both repair as well as pathological neovascularization in the eye have been observed in rodent models. This may be attributable to the particularly quiescent nature of the resident retinal vasculature (typical retinal EC turnover occurs every 4 years), thus facilitating the contribution of circulating cells to the newly forming vessel. In human–mouse chimeric models of ocular vascular injury, we observed that healthy human CD34⫹ cells effectively repair injured mouse/rat retina by directly participating in re-endothelialization of acellular capillaries (Figure 4).25

Cell-Based Therapies for Diabetic Complications

861

Figure 4. Human CD34ⴙ of nondiabetic, but not diabetic, origin integrate into degenerate vasculature in mouse eyes damaged by ischemia/reperfusion injury (Caballero et al25). Two days after intravitreal or systemic administration, integration of diabetic cells into existing vasculature was not observed (A), whereas cells of nondiabetic origin show extensive integration into small and medium sized vessels (yellow in the composite images) (B). Insets show separate red and green channels used to make the composite images. C, CD34⫹ cells home to an area of injury and traverse toward the ischemic region of the capillary (arrows).

Nephropathy Whereas the evidence for EPCs taking part in the pathogenesis of diabetic retinopathy is quite robust, the evidence for involvement in diabetic nephropathy is less strong, although a role for endothelial dysfunction has been documented in the development of diabetic nephropathy.114 In addition to the 3 different types of endothelium (vascular, peritubular, and glomerular), the kidney contains glomerular mesangial and tubular epithelial cells. Studies are yet to be carried out to identify stem/progenitor cell phenotypes that regenerate/ differentiate into one or more of these cell types. The participation of BMCs in kidney repair has been shown in different experimental studies115–117; however, these findings were challenged by a study using a chimeric mouse model.118 In addition, Lee et al119 observed homing of very few of the human mesenchymal stem cells (MSCs) in the glomeruli of STZ-diabetic NOD/SCID mice following intracardiac administration of the MSCs. More systematic studies need to be carried out to support the use of cell-based therapies in diabetic nephropathy.

Neuropathy Peripheral neuropathy is a common complication of diabetes, which may lead to foot ulcers that can result in amputation. Diabetes-induced impairment of the microcirculation precedes diabetic neuropathy. Therefore, re-endothelialization or vascular repair may in fact be essential for the restoration of nerve function. A few studies have tested the therapeutic potential of stem/progenitor cells in diabetic neuropathy. Using intramuscular injection of mononuclear cells derived from peripheral blood or BM, Hasegawa et al120 showed significant amelioration of diabetic neuropathy in rats with 4 weeks of STZ-diabetes. This study further showed this beneficial effect was not accompanied by angiogenesis but was blocked by treatment with VEGF-neutralizing antibody.

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

862

Circulation Research

March 19, 2010

In rats with 6 weeks of STZ-diabetes, Naruse et al121 observed amelioration of neuropathy associated with improved angiogenesis following intramuscular administration of human umbilical cord blood-derived 7-day culture-expanded EPCs. Recent study in rats with 12-weeks of STZ-diabetes treated with rat BMCs reported restoration of nerve function that was accompanied by revascularization but colocalization of injected cells with ECs was not observed.122 Jeong et al123 showed that intramuscular injection of murine BM-derived culture-expanded EPCs restored physiological responses and neural vascularity as well as transdifferentiation into ECs was observed in mice with 12 weeks of STZ-diabetes. This study also showed evidence for the durability of engrafted EPCs for ⬎12 months. Together, these studies suggest that the cell therapy as an innovative option for the treatment of diabetic neuropathy; however, no studies involving use of autologous diabetic cells have been reported.

Diabetic Macrovascular Complications Diabetes with or without clinical obesity is a high risk factor for the progression of atherosclerosis, which, in turn, manifests as coronary artery disease or peripheral vascular disease and recent studies observed close association between EPCs and incidence of atherosclerosis.56,124,125 It is not clear whether EPCs promote or alleviate the progression and severity of atherosclerosis. Controversial results from experimental studies could be attributable to the lack of uniformity in the age and gender of the animals used because these 2 factors seem to influence the impact of BMCs on the development of atherosclerosis.125 Early case reports suggested that BMCs may worsen restenosis; however, later studies showed no evidence for this outcome.126,127 George et al128 evaluated the effects of intravenous injection of BMCs or spleen-derived culture-expanded EPCs in apolipoprotein E knockout mice. Treatment with BMCs increased aortic sinus lesion size with increased plaque area; whereas treatment with EPCs resulted in an increased lipid core of the plaque with thinner fibrous cap and increased number of infiltrating CD3 cells. Both treatments resulted in decreased interleukin-10 levels, and, in addition, interleukin-6 and monocyte chemoattractant protein-1 were seen in the group that received BMCs. In a cholesterol-fed rabbit model of neointimal formation and inflammation, balloon-denuded and radiated iliac arteries were evaluated following treatment with peripheral blood EPCs or BMCs. No signs of decrease in neointimal thickening were observed after treatment with either population of cells twice at one and 2 weeks after denudation and radiation of arteries,129 whereas the study by Ma et al,130 in a similar rabbit model with denuded common carotid artery, reported significant decrease in stenosis by 2 weeks after treatment with EPCs and beneficial effect remained similar up to 15 weeks. The question that is yet to be clarified is which model is best as a representative model of human atherosclerosis, keeping in mind that not all studies in humans result in clear outcomes. A recent study by Subramaniyam et al131 in patients with peripheral artery disease showed beneficial effects of mobilization of BMCs with GM-CSF. Numbers of leukocytes and CD34⫹ cells were significantly increased by 2 weeks of

GM-CSF treatment, with an increased number of CFUs, suggesting that the mobilized cells are functional. Improved endothelial function and increased exercise capacity were observed 12 weeks after GM-CSF treatment. In contrast, the study by Horie et al132 using autologous implantation of mobilized peripheral blood mononuclear cells was inconclusive, and this was attributed to low numbers of CD34⫹ cells and decreased survival attributable to reasons other than peripheral limb ischemia. Significant progress has been accomplished in applying cell-based therapies in patients with coronary artery disease or myocardial ischemia and different ongoing clinical trials evaluated and confirmed the safety of cell-based therapies.41,42,133

Wound Healing and Ulcers Effective wound healing is an orchestrated response involving angiogenesis, enhanced cellularity, reepithelialization, and glandularization, which is indicative of cutaneous regeneration. Specificity of cellular phenotypes governing these different events is not yet known, but the phenomenon of wound healing is severely impaired in diabetes. Earlier studies in cell-based therapies for diabetic wound healing were focused on using fibroblasts; however, recent preclinical studies explored the benefits of BMCs. Healthy, nondiabetic CD34⫹ cells were shown to accelerate vascularization and wound healing in diabetic mice23; murine diabetic BMCs inhibited angiogenesis but produced wound healing in diabetic mice suggesting that these 2 events involve distinct mechanisms perhaps involving distinct cellular phenotypes.54,134 Murine BM-derived MSCs or their conditioned medium were shown to enhance wound healing with cutaneous regeneration that was not observed with CD34⫹ cells.135,136 BM stromal cells also have been shown to improve wound closure following topical or systemic administration in diabetic rats.137 Human fetal-derived CD133⫹ cells produced effective wound healing in diabetic mice.48 Recently, murine lin⫺ cells were found to be effective in diabetic wound healing and were shown to remain in wound areas for up to 28 days compared to lin⫹ cells that were not found after 7 days.138 These studies suggest that healthy progenitors may serve to enhance wound healing in diabetic patients.

Diabetic Erectile Dysfunction Erectile response is a neurovascular response involves both central and peripheral components. The peripheral component of this response is known to be impaired in diabetes and has been considered as a risk factor and a predictor of future cardiovascular events, suggesting that the diabetic impairment of erectile response precedes coronary artery disease. Experimental studies were reported showing the potential of cell-based therapy in correcting erectile dysfunction; however, to date, there are no studies reported in diabetic erectile dysfunction. Immortalized neural crest stem cells, K10 cells, transplanted in rat penile corpus cavernosum were shown to differentiate into cells with both EC- and smooth muscle cell (SMC)-specific markers.139 Human MSCs transduced with retroviral vector encoding v-myc were shown to differentiate

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

Jarajapu and Grant into endothelial and SMCs.140 However, these 2 studies have not evaluated functional/erectile responses. In a rat model of age-associated erectile dysfunction, Bivalacqua et al141 studied erectile responses in rat at 7 and 21 days after intracorporeal injection of ex vivo– expanded rat MSCs that were or were not transduced with adenoviral vector encoding eNOS. Functional responses were shown to be improved in 21 days, but not in 7 days. The injected cells expressed EC- and SMC-specific markers at both the 7 and 21 day time points.

Autologous Cell Therapy in Diabetic Vascular Complications Despite strong lines of evidence supporting the concept of therapeutic angiogenesis by putative EPCs, few studies have used diabetic models, particularly, testing autologous transplantation. As described above, diabetic cells have impaired reparative function with complex underlying mechanisms. Cells for treatment of diabetic vasculopathy should be equipped with cellular and molecular armamentarium to withstand the in vivo diabetic microenvironment. Their migration to the site of repair and the ability to transmigrate, differentiate, and engraft into a blood vessel should be minimally affected by the diabetic environment. Several approaches have been proposed, but few have been tried in diabetic cells and evaluated in diabetic models. Hypoxic preconditioning of EPCs was shown to enhance their angiogenic potential.142 However, it is questionable whether diabetic EPCs would be as responsive as normal cells to hypoxia.

Cell-Based Therapies for Diabetic Complications

863

EPC reparative function after diabetes-related endothelial injury.107

Ex Vivo Manipulation of Diabetic Cells Genetic Modification of Cells Accumulating evidence indicates that biologically modified EPCs to release proangiogenic factors may be a potent therapeutic tool compared to unmodified cells. In this regard, Iwaguro et al143 transduced murine EPCs ex vivo with adenoviral vector encoding VEGF gene and demonstrated that VEGF-expressing cells had enhanced angiogenic potential in in vitro and in in vivo assays. Murasawa et al144 have used a similar approach to delay human EPC senescence by transducing progenitor cells with hTERT (human telomerase reverse transcriptase) and observed increased capillary density and blood flow in the mouse hindlimb ischemia. EPCs that were transduced to overexpress eNOS were shown to inhibit neointimal hyperplasia in a rabbit model of carotid artery balloon angioplasty.145 Choi et al146 have targeted glycogen synthase kinase-3␤ in human early EPCs and late outgrowth EPCs and reported that this genetic modification enhanced their vasoregenerative potential in mouse hindlimb ischemia. Human MSCs transduced with the prosurvival gene Akt-1 showed enhanced viability and greater functional repair in a mouse infarct model.147 We recently identified that expression of IGFBP-3 (IGF-1 binding protein-3) in stem/ progenitor cells or in the area of vascular injury dramatically enhanced the homing and vascular repair.112

Ex Vivo Treatment With Small Molecules

Neovascularization: When Is There Too Little or Too Much? Perhaps one of the most perplexing aspects of diabetes vascular dysfunction is how the retina can respond to the same insults with pathological neovascularization and the other vascular beds consistently demonstrate reduced angiogenesis. This “paradox” remains at the center of key questions in the field. So how can this be reconciled? One explanation is that the diabetic retina and vitreous represents a unique environment in which high concentrations of complementary growth factors (VEGF, insulin-like growth factor [IGF]-1, fibroblast growth factor, SDF-1, erythropoietin) act in a synergistic manner in contiguous environments (retina and vitreous) that provide scaffolding (remnant “hyaloid”) for new vessel formation. This unique structural arrangement and an accompanying plethora of inflammatory cells likely set up an ideal environment for neovessel formation. Once the damage is widespread, inflammation increases and chemokines are produced by the ischemic retina; the BM responds by increasing the production of EPCs, which in the pathological setting of the growth factor rich vitreous and retina may result in pathological neovascularization. Thus, the cooperation of circulating EPCs with resident retinal vasculature and circulating inflammatory monocytes results in “too much” neovascularization, leading to PDR. In contrast, in nonocular vascular beds, the level of neovascularization is markedly reduced, in part, because of progressive basement membrane modification by AGEs, which contributes to impairment of

Ex vivo treatment of BMCs with a novel molecule, AVE9488, that increases eNOS expression by stimulating eNOS promoter activity, enhanced the angiogenic function of EPCs in mouse hindlimb ischemia.148 Similar effects were produced by ex vivo treatment with IGF-1, which indeed reversed age-associated decrease in human and murine EPCs.149 However, one should be very cautious when adopting this approach in diabetic cells, because without correcting the diabetic oxidative environment, an increase in NO production by iNOS may result in higher production of reactive nitrogen species that further increase oxidative stress. Sphingosine-1-phosphate and its synthetic equivalent, FTY720, were shown to stimulate EPC function in restoring blood flow in mouse ischemic limbs via activating CXCR4coupled signaling pathway.150 Active p38 MAPK downregulates the number of BMCs in response to high glucose or tumor necrosis factor-␣ and ex vivo treatment with the p38 MAPK inhibitor SB203580 has been shown to increase the proliferation of EPCs that can be used for transplantation.151 Sorrentino et al152 showed that treatment of human diabetic EPCs with the peroxisome proliferator-activated receptor-␥ agonist rosiglitazone improved their angiogenic function in a mouse model of carotid injury by reducing NADPH oxidase– dependent ROS production and enhancing bioavailable NO levels. Agents or molecular maneuvers that can reduce oxidative stress as well as increase eNOS expression/activation would be of immense value in correcting diabetic EPC dysfunction.

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

864

Circulation Research

March 19, 2010

In Vivo Manipulation of Diabetic Cells In vivo manipulation of EPCs may involve modifying the diabetic milieu or the EPC niches to release functioning cells for homing to the areas of repair; however, possible options are very few. Treatment with growth hormone has been shown to reverse age-associated impairment of EPC function with increased eNOS expression, and this treatment resulted in elevated IGF-1 levels in mice and humans.149 Although mobilization of BM cells using G-CSF for therapeutic angiogenesis requires more thorough safety evaluation,69,70 studies using AMD3100 appear to be encouraging.153 AMD3100 is a small molecule antagonist of chemokine receptor CXCR4 and has been shown to mobilize BM cells in mice and humans.154 –156 In vivo treatment with AMD3100 or local administration of cells mobilized by AMD3100 accelerates blood flow to ischemic limbs in 12 week STZ-diabetic mouse.153 However, these studies need to be carried out in mice with different durations of diabetes, because it has been hypothesized that BM microenvironment may initially provide protection to resident EPCs from adverse diabetic milieu of the circulation and cells become dysfunctional only with longer durations of diabetes (Grant, MB, unpublished). Another CXCR4 antagonist, SDF-1␤P2G, has better angiogenic function compared to AMD3100 but has not yet been studied in diabetic models.157

Manipulation of Diabetic Host Environment A successful outcome of the cell-based therapy also depends on the host environment. An optimal “fertile” environment is essential for the adhesion and transmigration of cells that require interaction of cell surface molecules with extracellular matrix proteins. The oxidative diabetic milieu modifies the host environment, and the manipulation of which is required to enhance the reparative function of cells. Treatment with antioxidants or anti-AGE agents is an option. Treatment with insulin, glitazones, or statins may directly or indirectly makes the host environment favorable for the therapeutic behavior of the cells.

Future of Cell-Based Therapies Attempting translational studies before understanding the full identity and characteristics of EPC phenotype is likely fraught with difficulties. Which cell type is appropriate for a particular complication is still a difficult question to be answered. Although the study by Tendera et al158 found no difference in the efficacy of unselected BMCs and CD34⫹CXCR4⫹ cells, preclinical studies and ongoing clinical trials support the use of CD34⫹ cells compared to the total mononuclear leukocytes or unselected BMCs, although a higher dose of CD34⫹ cells was shown to be less efficacious than lower doses.42,159,160 As described above, autologous cell therapy in diabetics requires the ex vivo modification of EPCs for a better angiogenic outcome regardless of the cell type chosen. At the same time, PDR needs treatments inhibiting new retinal blood vessel formation, including blocking EPC recruitment and engraftment therein. Conversely, other sites and disease conditions may actually need an increase of EPC activity toward tissue repair, eg, in myocardial ischemia or wound healing. The previous sections

have emphasized the role of NO in maintaining normal function of EPC/BMCs and ways to maintain balanced NO and ROS levels. All BMC populations may have a key role in the repair process, and it is likely that combinations of progenitor cells will be needed, as will the simultaneous optimization of the diabetic environment into which these cells will be placed. Such a complex approach will likely be needed to optimally treat diabetic patients with vascular complications.

Acknowledgments We thank Dr Lynn C. Shaw for the preparation of schematics for this manuscript.

Sources of Funding This work was supported by NIH grants EY007739 and EY012601 (to M.B.G.).

Disclosures None.

References 1. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:964 –967. 2. Stump MM, Jordan GL Jr, Debakey ME, Halpert B. Endothelium grown from circulating blood on isolated intravascular Dacron hub. Am J Pathol. 1963;43:361–367. 3. Wu MH, Shi Q, Wechezak AR, Clowes AW, Gordon IL, Sauvage LR. Definitive proof of endothelialization of a Dacron arterial prosthesis in a human being. J Vasc Surg. 1995;21:862– 867. 4. Risau W. Mechanisms of angiogenesis. Nature. 1997;386:671– 674. 5. Carmeliet P. Angiogenesis in life, disease and medicine. Nature. 2005; 438:932–936. 6. Gehling UM, Ergun S, Schumacher U, Wagener C, Pantel K, Otte M, Schuch G, Schafhausen P, Mende T, Kilic N, Kluge K, Schafer B, Hossfeld DK, Fiedler W. In vitro differentiation of endothelial cells from AC133-positive progenitor cells. Blood. 2000;95:3106 –3112. 7. Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, Williams M, Oz MC, Hicklin DJ, Witte L, Moore MA, Rafii S. Expression of VEGFR-2 and AC133 by circulating human CD34(⫹) cells identifies a population of functional endothelial precursors. Blood. 2000;95:952–958. 8. Fernandez Pujol B, Lucibello FC, Gehling UM, Lindemann K, Weidner N, Zuzarte ML, Adamkiewicz J, Elsasser HP, Muller R, Havemann K. Endothelial-like cells derived from human CD14 positive monocytes. Differentiation. 2000;65:287–300. 9. Schmeisser A, Garlichs CD, Zhang H, Eskafi S, Graffy C, Ludwig J, Strasser RH, Daniel WG. Monocytes coexpress endothelial and macrophagocytic lineage markers and form cord-like structures in Matrigel under angiogenic conditions. Cardiovasc Res. 2001;49:671– 680. 10. Awad O, Dedkov EI, Jiao C, Bloomer S, Tomanek RJ, Schatteman GC. Differential healing activities of CD34⫹ and CD14⫹ endothelial cell progenitors. Arterioscler Thromb Vasc Biol. 2006;26:758 –764. 11. Quirici N, Soligo D, Caneva L, Servida F, Bossolasco P, Deliliers GL. Differentiation and expansion of endothelial cells from human bone marrow CD133(⫹) cells. Br J Haematol. 2001;115:186 –194. 12. Loomans CJ, Wan H, de Crom R, van Haperen R, de Boer HC, Leenen PJ, Drexhage HA, Rabelink TJ, van Zonneveld AJ, Staal FJ. Angiogenic murine endothelial progenitor cells are derived from a myeloid bone marrow fraction and can be identified by endothelial NO synthase expression. Arterioscler Thromb Vasc Biol. 2006;26:1760 –1767. 13. Harraz M, Jiao C, Hanlon HD, Hartley RS, Schatteman GC. CD34blood-derived human endothelial cell progenitors. Stem Cells. 2001;19: 304 –312. 14. Shi Q, Rafii S, Wu MH, Wijelath ES, Yu C, Ishida A, Fujita Y, Kothari S, Mohle R, Sauvage LR, Moore MA, Storb RF, Hammond WP. Evidence for circulating bone marrow-derived endothelial cells. Blood. 1998;92:362–367. 15. Takahashi T, Kalka C, Masuda H, Chen D, Silver M, Kearney M, Magner M, Isner JM, Asahara T. Ischemia- and cytokine-induced mobi-

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

Jarajapu and Grant

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

lization of bone marrow-derived endothelial progenitor cells for neovascularization. Nat Med. 1999;5:434 – 438. Hill JM, Zalos G, Halcox JP, Schenke WH, Waclawiw MA, Quyyumi AA, Finkel T. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med. 2003;348:593– 600. Schmidt-Lucke C, Rossig L, Fichtlscherer S, Vasa M, Britten M, Kamper U, Dimmeler S, Zeiher AM. Reduced number of circulating endothelial progenitor cells predicts future cardiovascular events: proof of concept for the clinical importance of endogenous vascular repair. Circulation. 2005;111:2981–2987. Werner N, Kosiol S, Schiegl T, Ahlers P, Walenta K, Link A, Bohm M, Nickenig G. Circulating endothelial progenitor cells and cardiovascular outcomes. N Engl J Med. 2005;353:999 –1007. Fadini GP, Sartore S, Albiero M, Baesso I, Murphy E, Menegolo M, Grego F, Vigili de Kreutzenberg S, Tiengo A, Agostini C, Avogaro A. Number and function of endothelial progenitor cells as a marker of severity for diabetic vasculopathy. Arterioscler Thromb Vasc Biol. 2006; 26:2140 –2146. Fadini GP, Coracina A, Baesso I, Agostini C, Tiengo A, Avogaro A, de Kreutzenberg SV. Peripheral blood CD34⫹KDR⫹ endothelial progenitor cells are determinants of subclinical atherosclerosis in a middle-aged general population. Stroke. 2006;37:2277–2282. Fadini GP, Pagano C, Baesso I, Kotsafti O, Doro D, de Kreutzenberg SV, Avogaro A, Agostini C, Dorigo MT. Reduced endothelial progenitor cells and brachial artery flow-mediated dilation as evidence of endothelial dysfunction in ocular hypertension and primary open-angle glaucoma. Acta Ophthalmol. 2010;88:135–141. Case J, Mead LE, Bessler WK, Prater D, White HA, Saadatzadeh MR, Bhavsar JR, Yoder MC, Haneline LS, Ingram DA. Human CD34⫹AC133⫹VEGFR-2⫹ cells are not endothelial progenitor cells but distinct, primitive hematopoietic progenitors. Exp Hematol. 2007; 35:1109 –1118. Sivan-Loukianova E, Awad OA, Stepanovic V, Bickenbach J, Schatteman GC. CD34⫹ blood cells accelerate vascularization and healing of diabetic mouse skin wounds. J Vasc Res. 2003;40:368 –377. Madeddu P, Emanueli C, Pelosi E, Salis MB, Cerio AM, Bonanno G, Patti M, Stassi G, Condorelli G, Peschle C. Transplantation of low dose CD34⫹KDR⫹ cells promotes vascular and muscular regeneration in ischemic limbs. FASEB J. 2004;18:1737–1739. Caballero S, Sengupta N, Afzal A, Chang KH, Li Calzi S, Guberski DL, Kern TS, Grant MB. Ischemic vascular damage can be repaired by healthy, but not diabetic, endothelial progenitor cells. Diabetes. 2007; 56:960 –967. Schatteman GC, Hanlon HD, Jiao C, Dodds SG, Christy BA. Bloodderived angioblasts accelerate blood-flow restoration in diabetic mice. J Clin Invest. 2000;106:571–578. Popa ER, Harmsen MC, Tio RA, van der Strate BW, Brouwer LA, Schipper M, Koerts J, De Jongste MJ, Hazenberg A, Hendriks M, van Luyn MJ. Circulating CD34⫹ progenitor cells modulate host angiogenesis and inflammation in vivo. J Mol Cell Cardiol. 2006;41:86 –96. Ziegelhoeffer T, Fernandez B, Kostin S, Heil M, Voswinckel R, Helisch A, Schaper W. Bone marrow-derived cells do not incorporate into the adult growing vasculature. Circ Res. 2004;94:230 –238. Rohde E, Bartmann C, Schallmoser K, Reinisch A, Lanzer G, Linkesch W, Guelly C, Strunk D. Immune cells mimic the morphology of endothelial progenitor colonies in vitro. Stem Cells. 2007;25:1746 –1752. Yoder MC, Mead LE, Prater D, Krier TR, Mroueh KN, Li F, Krasich R, Temm CJ, Prchal JT, Ingram DA. Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals. Blood. 2007;109:1801–1809. Prokopi M, Pula G, Mayr U, Devue C, Gallagher J, Xiao Q, Boulanger CM, Westwood N, Urbich C, Willeit J, Steiner M, Breuss J, Xu Q, Kiechl S, Mayr M. Proteomic analysis reveals presence of platelet microparticles in endothelial progenitor cell cultures. Blood. 2009;114: 723–732. Hur J, Yoon CH, Kim HS, Choi JH, Kang HJ, Hwang KK, Oh BH, Lee MM, Park YB. Characterization of two types of endothelial progenitor cells and their different contributions to neovasculogenesis. Arterioscler Thromb Vasc Biol. 2004;24:288 –293. Hristov M, Weber C. Endothelial progenitor cells: characterization, pathophysiology, and possible clinical relevance. J Cell Mol Med. 2004; 8:498 –508. Schatteman GC, Awad O. In vivo and in vitro properties of CD34⫹ and CD14⫹ endothelial cell precursors. Adv Exp Med Biol. 2003;522:9 –16.

Cell-Based Therapies for Diabetic Complications

865

35. Sieveking DP, Buckle A, Celermajer DS, Ng MK. Strikingly different angiogenic properties of endothelial progenitor cell subpopulations: insights from a novel human angiogenesis assay. J Am Coll Cardiol. 2008;51:660 – 668. 36. Schofield R. The stem cell system. Biomed Pharmacother. 1983;37: 375–380. 37. Guven H, Shepherd RM, Bach RG, Capoccia BJ, Link DC. The number of endothelial progenitor cell colonies in the blood is increased in patients with angiographically significant coronary artery disease. J Am Coll Cardiol. 2006;48:1579 –1587. 38. George J, Goldstein E, Abashidze S, Deutsch V, Shmilovich H, Finkelstein A, Herz I, Miller H, Keren G. Circulating endothelial progenitor cells in patients with unstable angina: association with systemic inflammation. Eur Heart J. 2004;25:1003–1008. 39. Hirata K, Li TS, Nishida M, Ito H, Matsuzaki M, Kasaoka S, Hamano K. Autologous bone marrow cell implantation as therapeutic angiogenesis for ischemic hindlimb in diabetic rat model. Am J Physiol Heart Circ Physiol. 2003;284:H66 –H70. 40. Urbich C, Dimmeler S. Endothelial progenitor cells: characterization and role in vascular biology. Circ Res. 2004;95:343–353. 41. Ward MR, Stewart DJ, Kutryk MJ. Endothelial progenitor cell therapy for the treatment of coronary disease, acute MI, and pulmonary arterial hypertension: current perspectives. Catheter Cardiovasc Interv. 2007; 70:983–998. 42. Sekiguchi H, Ii M, Losordo DW. The relative potency and safety of endothelial progenitor cells and unselected mononuclear cells for recovery from myocardial infarction and ischemia. J Cell Physiol. 2009; 219:235–242. 43. Krenning G, van Luyn MJ, Harmsen MC. Endothelial progenitor cell-based neovascularization: implications for therapy. Trends Mol Med. 2009;15:180 –189. 44. Majka M, Janowska-Wieczorek A, Ratajczak J, Ehrenman K, Pietrzkowski Z, Kowalska MA, Gewirtz AM, Emerson SG, Ratajczak MZ. Numerous growth factors, cytokines, and chemokines are secreted by human CD34(⫹) cells, myeloblasts, erythroblasts, and megakaryoblasts and regulate normal hematopoiesis in an autocrine/paracrine manner. Blood. 2001;97:3075–3085. 45. Krenning G, van der Strate B, Schipper M, Gallego YvSX, Fernandes B, van Luyn M, Harmsen M CD34(⫹) Cells Augment Endothelial Cell Differentiation of CD14(⫹) Endothelial Progenitor Cells in vitro. J Cell Mol Med. 2008. 46. Rehman J, Li J, Orschell CM, March KL. Peripheral blood “endothelial progenitor cells” are derived from monocyte/macrophages and secrete angiogenic growth factors. Circulation. 2003;107:1164 –1169. 47. Urbich C, Aicher A, Heeschen C, Dernbach E, Hofmann WK, Zeiher AM, Dimmeler S. Soluble factors released by endothelial progenitor cells promote migration of endothelial cells and cardiac resident progenitor cells. J Mol Cell Cardiol. 2005;39:733–742. 48. Barcelos LS, Duplaa C, Krankel N, Graiani G, Invernici G, Katare R, Siragusa M, Meloni M, Campesi I, Monica M, Simm A, Campagnolo P, Mangialardi G, Stevanato L, Alessandri G, Emanueli C, Madeddu P. Human CD133⫹ progenitor cells promote the healing of diabetic ischemic ulcers by paracrine stimulation of angiogenesis and activation of Wnt signaling. Circ Res. 2009;104:1095–1102. 49. Gnecchi M, He H, Melo LG, Noiseaux N, Morello F, de Boer RA, Zhang L, Pratt RE, Dzau VJ, Ingwall JS. Early beneficial effects of bone marrow-derived mesenchymal stem cells overexpressing Akt on cardiac metabolism after myocardial infarction. Stem Cells. 2009;27:971–979. 50. Burchfield JS, Dimmeler S. Role of paracrine factors in stem and progenitor cell mediated cardiac repair and tissue fibrosis. Fibrogenesis Tissue Repair. 2008;1:4. 51. He T, Peterson TE, Katusic ZS. Paracrine mitogenic effect of human endothelial progenitor cells: role of interleukin-8. Am J Physiol Heart Circ Physiol. 2005;289:H968 –H972. 52. Santhanam AV, Smith LA, He T, Nath KA, Katusic ZS. Endothelial progenitor cells stimulate cerebrovascular production of prostacyclin by paracrine activation of cyclooxygenase-2. Circ Res. 2007;100: 1379 –1388. 53. He T, Lu T, d’Uscio LV, Lam CF, Lee HC, Katusic ZS. Angiogenic function of prostacyclin biosynthesis in human endothelial progenitor cells. Circ Res. 2008;103:80 – 88. 54. Awad O, Jiao C, Ma N, Dunnwald M, Schatteman GC. Obese diabetic mouse environment differentially affects primitive and monocytic endothelial cell progenitors. Stem Cells. 2005;23:575–583.

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

866

Circulation Research

March 19, 2010

55. Jarajapu YP, Thampi P, Caballero S, Boulton M, Grant MB. Differential paracrine dysfunction and hypoxic desensitization in diabetic CD34⫹ and CD14⫹ bone marrow derived cells (BMDC). Circulation. 2009; 120:S1124. 56. Fadini GP, Miorin M, Facco M, Bonamico S, Baesso I, Grego F, Menegolo M, de Kreutzenberg SV, Tiengo A, Agostini C, Avogaro A. Circulating endothelial progenitor cells are reduced in peripheral vascular complications of type 2 diabetes mellitus. J Am Coll Cardiol. 2005;45:1449 –1457. 57. Fadini GP, Pucci L, Vanacore R, Baesso I, Penno G, Balbarini A, Di Stefano R, Miccoli R, de Kreutzenberg S, Coracina A, Tiengo A, Agostini C, Del Prato S, Avogaro A. Glucose tolerance is negatively associated with circulating progenitor cell levels. Diabetologia. 2007; 50:2156 –2163. 58. Fadini GP, Sartore S, Agostini C, Avogaro A. Significance of endothelial progenitor cells in subjects with diabetes. Diabetes Care. 2007;30: 1305–1313. 59. Egan CG, Lavery R, Caporali F, Fondelli C, Laghi-Pasini F, Dotta F, Sorrentino V. Generalised reduction of putative endothelial progenitors and CXCR4-positive peripheral blood cells in type 2 diabetes. Diabetologia. 2008;51:1296 –1305. 60. Tepper OM, Galiano RD, Capla JM, Kalka C, Gagne PJ, Jacobowitz GR, Levine JP, Gurtner GC. Human endothelial progenitor cells from type II diabetics exhibit impaired proliferation, adhesion, and incorporation into vascular structures. Circulation. 2002;106:2781–2786. 61. Loomans CJ, de Koning EJ, Staal FJ, Rookmaaker MB, Verseyden C, de Boer HC, Verhaar MC, Braam B, Rabelink TJ, van Zonneveld AJ. Endothelial progenitor cell dysfunction: a novel concept in the pathogenesis of vascular complications of type 1 diabetes. Diabetes. 2004; 53:195–199. 62. Lee IG, Chae SL, Kim JC. Involvement of circulating endothelial progenitor cells and vasculogenic factors in the pathogenesis of diabetic retinopathy. Eye (Lond). 2006;20:546 –552. 63. Kusuyama T, Omura T, Nishiya D, Enomoto S, Matsumoto R, Takeuchi K, Yoshikawa J, Yoshiyama M. Effects of treatment for diabetes mellitus on circulating vascular progenitor cells. J Pharmacol Sci. 2006; 102:96 –102. 64. Fadini GP, Sartore S, Schiavon M, Albiero M, Baesso I, Cabrelle A, Agostini C, Avogaro A. Diabetes impairs progenitor cell mobilisation after hindlimb ischaemia-reperfusion injury in rats. Diabetologia. 2006; 49:3075–3084. 65. Yan J, Tie G, Park B, Yan Y, Nowicki PT, Messina LM. Recovery from hind limb ischemia is less effective in type 2 than in type 1 diabetic mice: Roles of endothelial nitric oxide synthase and endothelial progenitor cells. J Vasc Surg. 2009;50:1412–1422. 66. Nguyen TQ, Chon H, van Nieuwenhoven FA, Braam B, Verhaar MC, Goldschmeding R. Myofibroblast progenitor cells are increased in number in patients with type 1 diabetes and express less bone morphogenetic protein 6: a novel clue to adverse tissue remodelling? Diabetologia. 2006;49:1039 –1048. 67. Busik JV, Tikhonenko M, Bhatwadekar A, Opreanu M, Yakubova N, Caballero S, Player D, Nakagawa T, Afzal A, Kielczewski J, Sochacki A, Hasty S, Li Calzi S, Kim S, Duclas SK, Segal MS, Guberski DL, Esselman WJ, Boulton ME, Grant MB. Diabetic retinopathy is associated with bone marrow neuropathy and a depressed peripheral clock. J Exp Med. 2009;206:2897–2906. 68. Humpert PM, Neuwirth R, Battista MJ, Voronko O, von Eynatten M, Konrade I, Rudofsky G Jr, Wendt T, Hamann A, Morcos M, Nawroth PP, Bierhaus A. SDF-1 genotype influences insulin-dependent mobilization of adult progenitor cells in type 2 diabetes. Diabetes Care. 2005;28:934 –936. 69. Honold J, Lehmann R, Heeschen C, Walter DH, Assmus B, Sasaki K, Martin H, Haendeler J, Zeiher AM, Dimmeler S. Effects of granulocyte colony simulating factor on functional activities of endothelial progenitor cells in patients with chronic ischemic heart disease. Arterioscler Thromb Vasc Biol. 2006;26:2238 –2243. 70. Kang HJ, Kim HS, Zhang SY, Park KW, Cho HJ, Koo BK, Kim YJ, Soo Lee D, Sohn DW, Han KS, Oh BH, Lee MM, Park YB. Effects of intracoronary infusion of peripheral blood stem-cells mobilised with granulocyte-colony stimulating factor on left ventricular systolic function and restenosis after coronary stenting in myocardial infarction: the MAGIC cell randomised clinical trial. Lancet. 2004;363:751–756. 71. Segal MS, Shah R, Afzal A, Perrault CM, Chang K, Schuler A, Beem E, Shaw LC, Li Calzi S, Harrison JK, Tran-Son-Tay R, Grant MB. Nitric oxide cytoskeletal-induced alterations reverse the endothelial progenitor

72.

73.

74.

75. 76.

77.

78.

79.

80.

81.

82.

83. 84.

85.

86.

87.

88.

89.

90.

cell migratory defect associated with diabetes. Diabetes. 2006;55: 102–109. Tamarat R, Silvestre JS, Le Ricousse-Roussanne S, Barateau V, Lecomte-Raclet L, Clergue M, Duriez M, Tobelem G, Levy BI. Impairment in ischemia-induced neovascularization in diabetes: bone marrow mononuclear cell dysfunction and therapeutic potential of placenta growth factor treatment. Am J Pathol. 2004;164:457– 466. Kaesemeyer WH, Ogonowski AA, Jin L, Caldwell RB, Caldwell RW. Endothelial nitric oxide synthase is a site of superoxide synthesis in endothelial cells treated with glyceryl trinitrate. Br J Pharmacol. 2000; 131:1019 –1023. Berkowitz DE, White R, Li D, Minhas KM, Cernetich A, Kim S, Burke S, Shoukas AA, Nyhan D, Champion HC, Hare JM. Arginase reciprocally regulates nitric oxide synthase activity and contributes to endothelial dysfunction in aging blood vessels. Circulation. 2003;108: 2000 –2006. Baylis C. Arginine, arginine analogs and nitric oxide production in chronic kidney disease. Nat Clin Pract Nephrol. 2006;2:209 –220. Ellis EA, Guberski DL, Hutson B, Grant MB. Time course of NADH oxidase, inducible nitric oxide synthase and peroxynitrite in diabetic retinopathy in the BBZ/WOR rat. Nitric Oxide. 2002;6:295–304. Szabo C, Mabley JG, Moeller SM, Shimanovich R, Pacher P, Virag L, Soriano FG, Van Duzer JH, Williams W, Salzman AL, Groves JT. Part I: pathogenetic role of peroxynitrite in the development of diabetes and diabetic vascular complications: studies with FP15, a novel potent peroxynitrite decomposition catalyst. Mol Med. 2002;8:571–580. Kobayashi T, Taguchi K, Takenouchi Y, Matsumoto T, Kamata K. Insulin-induced impairment via peroxynitrite production of endothelium-dependent relaxation and sarco/endoplasmic reticulum Ca(2⫹)ATPase function in aortas from diabetic rats. Free Radic Biol Med. 2007;43:431– 443. Camici GG, Schiavoni M, Francia P, Bachschmid M, Martin-Padura I, Hersberger M, Tanner FC, Pelicci P, Volpe M, Anversa P, Luscher TF, Cosentino F. Genetic deletion of p66(Shc) adaptor protein prevents hyperglycemia-induced endothelial dysfunction and oxidative stress. Proc Natl Acad Sci U S A. 2007;104:5217–5222. Leal EC, Manivannan A, Hosoya K, Terasaki T, Cunha-Vaz J, Ambrosio AF, Forrester JV. Inducible nitric oxide synthase isoform is a key mediator of leukostasis and blood-retinal barrier breakdown in diabetic retinopathy. Invest Ophthalmol Vis Sci. 2007;48:5257–5265. Nagareddy PR, Xia Z, McNeill JH, MacLeod KM. Increased expression of iNOS is associated with endothelial dysfunction and impaired pressor responsiveness in streptozotocin-induced diabetes. Am J Physiol Heart Circ Physiol. 2005;289:H2144 –H2152. Cheng X, Cheng XS, Kuo KH, Pang CC. Inhibition of iNOS augments cardiovascular action of noradrenaline in streptozotocin-induced diabetes. Cardiovasc Res. 2004;64:298 –307. Aicher A, Zeiher AM, Dimmeler S. Mobilizing endothelial progenitor cells. Hypertension. 2005;45:321–325. Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR, Crystal RG, Besmer P, Lyden D, Moore MA, Werb Z, Rafii S. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell. 2002;109:625– 637. Shintani S, Murohara T, Ikeda H, Ueno T, Honma T, Katoh A, Sasaki K, Shimada T, Oike Y, Imaizumi T. Mobilization of endothelial progenitor cells in patients with acute myocardial infarction. Circulation. 2001;103:2776 –2779. Thum T, Fraccarollo D, Schultheiss M, Froese S, Galuppo P, Widder JD, Tsikas D, Ertl G, Bauersachs J. Endothelial nitric oxide synthase uncoupling impairs endothelial progenitor cell mobilization and function in diabetes. Diabetes. 2007;56:666 – 674. Vasa M, Breitschopf K, Zeiher AM, Dimmeler S. Nitric oxide activates telomerase and delays endothelial cell senescence. Circ Res. 2000;87: 540 –542. Kuki S, Imanishi T, Kobayashi K, Matsuo Y, Obana M, Akasaka T. Hyperglycemia accelerated endothelial progenitor cell senescence via the activation of p38 mitogen-activated protein kinase. Circ J. 2006;70: 1076 –1081. Rosso A, Balsamo A, Gambino R, Dentelli P, Falcioni R, Cassader M, Pegoraro L, Pagano G, Brizzi MF. p53 Mediates the accelerated onset of senescence of endothelial progenitor cells in diabetes. J Biol Chem. 2006;281:4339 – 4347. Balestrieri ML, Rienzo M, Felice F, Rossiello R, Grimaldi V, Milone L, Casamassimi A, Servillo L, Farzati B, Giovane A, Napoli C. High

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

Jarajapu and Grant

91.

92.

93.

94.

95.

96.

97.

98.

99.

100.

101.

102.

103.

104.

105.

106.

107.

108.

109.

glucose downregulates endothelial progenitor cell number via SIRT1. Biochim Biophys Acta. 2008;1784:936 –945. Loomans CJ, van Haperen R, Duijs JM, Verseyden C, de Crom R, Leenen PJ, Drexhage HA, de Boer HC, de Koning EJ, Rabelink TJ, Staal FJ, van Zonneveld AJ. Differentiation of bone marrow-derived endothelial progenitor cells is shifted into a proinflammatory phenotype by hyperglycemia. Mol Med. 2009;15:152–159. Ushio-Fukai M, Urao N. Novel role of NADPH oxidase in angiogenesis and stem/progenitor cell function. Antioxid Redox Signal. 2009;11: 2517–2533. Hink U, Li H, Mollnau H, Oelze M, Matheis E, Hartmann M, Skatchkov M, Thaiss F, Stahl RA, Warnholtz A, Meinertz T, Griendling K, Harrison DG, Forstermann U, Munzel T. Mechanisms underlying endothelial dysfunction in diabetes mellitus. Circ Res. 2001;88:e14 – e22. Guzik TJ, Mussa S, Gastaldi D, Sadowski J, Ratnatunga C, Pillai R, Channon KM. Mechanisms of increased vascular superoxide production in human diabetes mellitus: role of NAD(P)H oxidase and endothelial nitric oxide synthase. Circulation. 2002;105:1656 –1662. Piccoli C, D’Aprile A, Ripoli M, Scrima R, Lecce L, Boffoli D, Tabilio A, Capitanio N. Bone-marrow derived hematopoietic stem/progenitor cells express multiple isoforms of NADPH oxidase and produce constitutively reactive oxygen species. Biochem Biophys Res Commun. 2007; 353:965–972. Jang YY, Sharkis SJ. A low level of reactive oxygen species selects for primitive hematopoietic stem cells that may reside in the low-oxygenic niche. Blood. 2007;110:3056 –3063. Urao N, Inomata H, Razvi M, Kim HW, Wary K, McKinney R, Fukai T, Ushio-Fukai M. Role of nox2-based NADPH oxidase in bone marrow and progenitor cell function involved in neovascularization induced by hindlimb ischemia. Circ Res. 2008;103:212–220. Ito K, Hirao A, Arai F, Takubo K, Matsuoka S, Miyamoto K, Ohmura M, Naka K, Hosokawa K, Ikeda Y, Suda T. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat Med. 2006;12:446 – 451. Dernbach E, Urbich C, Brandes RP, Hofmann WK, Zeiher AM, Dimmeler S. Antioxidative stress-associated genes in circulating progenitor cells: evidence for enhanced resistance against oxidative stress. Blood. 2004;104:3591–3597. He T, Peterson TE, Holmuhamedov EL, Terzic A, Caplice NM, Oberley LW, Katusic ZS. Human endothelial progenitor cells tolerate oxidative stress due to intrinsically high expression of manganese superoxide dismutase. Arterioscler Thromb Vasc Biol. 2004;24:2021–2027. Ingram DA, Krier TR, Mead LE, McGuire C, Prater DN, Bhavsar J, Saadatzadeh MR, Bijangi-Vishehsaraei K, Li F, Yoder MC, Haneline LS. Clonogenic endothelial progenitor cells are sensitive to oxidative stress. Stem Cells. 2007;25:297–304. Jarajapu YP, Verma A, Kent D, Li Q, Grant MB Inhibition of NADPH oxidase restores NO availability and migratory function in diabetic CD34 cells. FASEB J. 2009;23:937.2. Scheubel RJ, Kahrstedt S, Weber H, Holtz J, Friedrich I, Borgermann J, Silber RE, Simm A. Depression of progenitor cell function by advanced glycation endproducts (AGEs): potential relevance for impaired angiogenesis in advanced age and diabetes. Exp Gerontol. 2006;41:540 –548. Sun C, Liang C, Ren Y, Zhen Y, He Z, Wang H, Tan H, Pan X, Wu Z. Advanced glycation end products depress function of endothelial progenitor cells via p38 and ERK 1/2 mitogen-activated protein kinase pathways. Basic Res Cardiol. 2009;104:42– 49. Fujii H, Li SH, Szmitko PE, Fedak PW, Verma S. C-reactive protein alters antioxidant defenses and promotes apoptosis in endothelial progenitor cells. Arterioscler Thromb Vasc Biol. 2006;26:2476 –2482. Chen Q, Dong L, Wang L, Kang L, Xu B. Advanced glycation end products impair function of late endothelial progenitor cells through effects on protein kinase Akt and cyclooxygenase-2. Biochem Biophys Res Commun. 2009;381:192–197. Bhatwadekar AD, Glenn JV, Li G, Curtis TM, Gardiner TA, Stitt AW. Advanced glycation of fibronectin impairs vascular repair by endothelial progenitor cells: implications for vasodegeneration in diabetic retinopathy. Invest Ophthalmol Vis Sci. 2008;49:1232–1241. Negre-Salvayre A, Salvayre R, Auge N, Pamplona R, Portero-Otin M. Hyperglycemia and glycation in diabetic complications. Antioxid Redox Signal. 2009;11:3071–3109. Brunner S, Schernthaner GH, Satler M, Elhenicky M, Hoellerl F, Schmid-Kubista KE, Zeiler F, Binder S, Schernthaner G. Correlation of different circulating endothelial progenitor cells to stages of diabetic

Cell-Based Therapies for Diabetic Complications

110.

111.

112.

113.

114.

115.

116.

117.

118.

119.

120.

121.

122.

123.

124.

125.

126.

127.

867

retinopathy: first in vivo data. Invest Ophthalmol Vis Sci. 2009;50: 392–398. Grant MB, May WS, Caballero S, Brown GA, Guthrie SM, Mames RN, Byrne BJ, Vaught T, Spoerri PE, Peck AB, Scott EW. Adult hematopoietic stem cells provide functional hemangioblast activity during retinal neovascularization. Nat Med. 2002;8:607– 612. Ritter MR, Banin E, Moreno SK, Aguilar E, Dorrell MI, Friedlander M. Myeloid progenitors differentiate into microglia and promote vascular repair in a model of ischemic retinopathy. J Clin Invest. 2006;116: 3266 –3276. Kielczewski JL, Jarajapu YP, McFarland EL, Cai J, Afzal A, Calzi SL, Chang KH, Lydic T, Shaw LC, Busik J, Hughes J, Cardounel AJ, Wilson K, Lyons TJ, Boulton ME, Mames RN, Chan-Ling T, Grant MB. Insulin-like growth factor binding protein-3 mediates vascular repair by enhancing nitric oxide generation. Circ Res. 2009;105:897–905. Otani A, Dorrell MI, Kinder K, Moreno SK, Nusinowitz S, Banin E, Heckenlively J, Friedlander M. Rescue of retinal degeneration by intravitreally injected adult bone marrow-derived lineage-negative hematopoietic stem cells. J Clin Invest. 2004;114:765–774. Nakagawa T, Segal M, Croker B, Johnson RJ. A breakthrough in diabetic nephropathy: the role of endothelial dysfunction. Nephrol Dial Transplant. 2007;22:2775–2777. Poulsom R, Forbes SJ, Hodivala-Dilke K, Ryan E, Wyles S, Navaratnarasah S, Jeffery R, Hunt T, Alison M, Cook T, Pusey C, Wright NA. Bone marrow contributes to renal parenchymal turnover and regeneration. J Pathol. 2001;195:229 –235. Togel F, Hu Z, Weiss K, Isaac J, Lange C, Westenfelder C. Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms. Am J Physiol Renal Physiol. 2005;289:F31–F42. Ikarashi K, Li B, Suwa M, Kawamura K, Morioka T, Yao J, Khan F, Uchiyama M, Oite T. Bone marrow cells contribute to regeneration of damaged glomerular endothelial cells. Kidney Int. 2005;67:1925–1933. Duffield JS, Park KM, Hsiao LL, Kelley VR, Scadden DT, Ichimura T, Bonventre JV. Restoration of tubular epithelial cells during repair of the postischemic kidney occurs independently of bone marrow-derived stem cells. J Clin Invest. 2005;115:1743–1755. Lee RH, Seo MJ, Reger RL, Spees JL, Pulin AA, Olson SD, Prockop DJ. Multipotent stromal cells from human marrow home to and promote repair of pancreatic islets and renal glomeruli in diabetic NOD/scid mice. Proc Natl Acad Sci U S A. 2006;103:17438 –17443. Hasegawa T, Kosaki A, Shimizu K, Matsubara H, Mori Y, Masaki H, Toyoda N, Inoue-Shibata M, Nishikawa M, Iwasaka T. Amelioration of diabetic peripheral neuropathy by implantation of hematopoietic mononuclear cells in streptozotocin-induced diabetic rats. Exp Neurol. 2006; 199:274 –280. Naruse K, Hamada Y, Nakashima E, Kato K, Mizubayashi R, Kamiya H, Yuzawa Y, Matsuo S, Murohara T, Matsubara T, Oiso Y, Nakamura J. Therapeutic neovascularization using cord blood-derived endothelial progenitor cells for diabetic neuropathy. Diabetes. 2005;54:1823–1828. Kim H, Park JS, Choi YJ, Kim MO, Huh YH, Kim SW, Han JW, Lee J, Kim S, Houge MA, Ii M, Yoon YS. Bone marrow mononuclear cells have neurovascular tropism and improve diabetic neuropathy. Stem Cells. 2009;27:1686 –1696. Jeong JO, Kim MO, Kim H, Lee MY, Kim SW, Ii M, Lee JU, Lee J, Choi YJ, Cho HJ, Lee N, Silver M, Wecker A, Kim DW, Yoon YS. Dual angiogenic and neurotrophic effects of bone marrow-derived endothelial progenitor cells on diabetic neuropathy. Circulation. 2009;119: 699 –708. Goldschmidt-Clermont PJ. Loss of bone marrow-derived vascular progenitor cells leads to inflammation and atherosclerosis. Am Heart J. 2003;146:S5–S12. Rauscher FM, Goldschmidt-Clermont PJ, Davis BH, Wang T, Gregg D, Ramaswami P, Pippen AM, Annex BH, Dong C, Taylor DA. Aging, progenitor cell exhaustion, and atherosclerosis. Circulation. 2003;108: 457– 463. Meyer GP, Wollert KC, Lotz J, Steffens J, Lippolt P, Fichtner S, Hecker H, Schaefer A, Arseniev L, Hertenstein B, Ganser A, Drexler H. Intracoronary bone marrow cell transfer after myocardial infarction: eighteen months’ follow-up data from the randomized, controlled BOOST (BOne marrOw transfer to enhance ST-elevation infarct regeneration) trial. Circulation. 2006;113:1287–1294. Schachinger V, Assmus B, Britten MB, Honold J, Lehmann R, Teupe C, Abolmaali ND, Vogl TJ, Hofmann WK, Martin H, Dimmeler S, Zeiher AM. Transplantation of progenitor cells and regeneration enhancement

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

868

128.

129.

130.

131.

132.

133. 134.

135.

136.

137.

138.

139.

140.

141.

142.

143.

144.

145.

Circulation Research

March 19, 2010

in acute myocardial infarction: final one-year results of the TOPCARE-AMI Trial. J Am Coll Cardiol. 2004;44:1690 –1699. George J, Afek A, Abashidze A, Shmilovich H, Deutsch V, Kopolovich J, Miller H, Keren G. Transfer of endothelial progenitor and bone marrow cells influences atherosclerotic plaque size and composition in apolipoprotein E knockout mice. Arterioscler Thromb Vasc Biol. 2005; 25:2636 –2641. Waksman R, Baffour R, Pakala R, Scheinowitz M, Hellinga D, Seabron R, Chan R, Kolodgie F, Virmani R. Effects of exogenous peripheralblood-derived endothelial progenitor cells or unfractionated bonemarrow-derived cells on neointimal formation and inflammation in cholesterol-fed, balloon-denuded, and radiated iliac arteries of inbred rabbits. Cardiovasc Revasc Med. 2009;10:110 –116. Ma ZL, Mai XL, Sun JH, Ju SH, Yang X, Ni Y, Teng GJ. Inhibited atherosclerotic plaque formation by local administration of magnetically labeled endothelial progenitor cells (EPCs) in a rabbit model. Atherosclerosis. 2009;205:80 – 86. Subramaniyam V, Waller EK, Murrow JR, Manatunga A, Lonial S, Kasirajan K, Sutcliffe D, Harris W, Taylor WR, Alexander RW, Quyyumi AA. Bone marrow mobilization with granulocyte macrophage colony-stimulating factor improves endothelial dysfunction and exercise capacity in patients with peripheral arterial disease. Am Heart J. 2009; 158:53– 60.e1. Horie T, Onodera R, Akamastu M, Ichikawa Y, Hoshino J, Kaneko E, Iwashita C, Ishida A, Tsukamoto T, Teramukai S, Fukushima M, Kawamura A. Long-term clinical outcomes for patients with lower limb ischemia implanted with G-CSF-mobilized autologous peripheral blood mononuclear cells. Atherosclerosis. 2010;208:461– 466. Dimmeler S, Burchfield J, Zeiher AM. Cell-based therapy of myocardial infarction. Arterioscler Thromb Vasc Biol. 2008;28:208 –216. Stepanovic V, Awad O, Jiao C, Dunnwald M, Schatteman GC. Leprdb diabetic mouse bone marrow cells inhibit skin wound vascularization but promote wound healing. Circ Res. 2003;92:1247–1253. Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells. 2007;25:2648 –2659. Javazon EH, Keswani SG, Badillo AT, Crombleholme TM, Zoltick PW, Radu AP, Kozin ED, Beggs K, Malik AA, Flake AW. Enhanced epithelial gap closure and increased angiogenesis in wounds of diabetic mice treated with adult murine bone marrow stromal progenitor cells. Wound Repair Regen. 2007;15:350 –359. Kwon DS, Gao X, Liu YB, Dulchavsky DS, Danyluk AL, Bansal M, Chopp M, McIntosh K, Arbab AS, Dulchavsky SA, Gautam SC. Treatment with bone marrow-derived stromal cells accelerates wound healing in diabetic rats. Int Wound J. 2008;5:453– 463. Lin CD, Allori AC, Macklin JE, Sailon AM, Tanaka R, Levine JP, Saadeh PB, Warren SM. Topical lineage-negative progenitor-cell therapy for diabetic wounds. Plast Reconstr Surg. 2008;122:1341–1351. Song YS, Lee HJ, Park IH, Lim IS, Ku JH, Kim SU. Human neural crest stem cells transplanted in rat penile corpus cavernosum to repair erectile dysfunction. BJU Int. 2008;102:220 –224. Song YS, Lee HJ, Park IH, Kim WK, Ku JH, Kim SU. Potential differentiation of human mesenchymal stem cell transplanted in rat corpus cavernosum toward endothelial or smooth muscle cells. Int J Impot Res. 2007;19:378 –385. Bivalacqua TJ, Deng W, Kendirci M, Usta MF, Robinson C, Taylor BK, Murthy SN, Champion HC, Hellstrom WJ, Kadowitz PJ. Mesenchymal stem cells alone or ex vivo gene modified with endothelial nitric oxide synthase reverse age-associated erectile dysfunction. Am J Physiol Heart Circ Physiol. 2007;292:H1278 –H1290. Akita T, Murohara T, Ikeda H, Sasaki K, Shimada T, Egami K, Imaizumi T. Hypoxic preconditioning augments efficacy of human endothelial progenitor cells for therapeutic neovascularization. Lab Invest. 2003;83:65–73. Iwaguro H, Yamaguchi J, Kalka C, Murasawa S, Masuda H, Hayashi S, Silver M, Li T, Isner JM, Asahara T. Endothelial progenitor cell vascular endothelial growth factor gene transfer for vascular regeneration. Circulation. 2002;105:732–738. Murasawa S, Llevadot J, Silver M, Isner JM, Losordo DW, Asahara T. Constitutive human telomerase reverse transcriptase expression enhances regenerative properties of endothelial progenitor cells. Circulation. 2002; 106:1133–1139. Kong D, Melo LG, Mangi AA, Zhang L, Lopez-Ilasaca M, Perrella MA, Liew CC, Pratt RE, Dzau VJ. Enhanced inhibition of neointimal hyperplasia

146.

147.

148.

149.

150.

151.

152.

153.

154.

155.

156.

157.

158.

159.

by genetically engineered endothelial progenitor cells. Circulation. 2004; 109:1769 –1775. Choi JH, Hur J, Yoon CH, Kim JH, Lee CS, Youn SW, Oh IY, Skurk C, Murohara T, Park YB, Walsh K, Kim HS. Augmentation of therapeutic angiogenesis using genetically modified human endothelial progenitor cells with altered glycogen synthase kinase-3beta activity. J Biol Chem. 2004;279:49430 – 49438. Mangi AA, Noiseux N, Kong D, He H, Rezvani M, Ingwall JS, Dzau VJ. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat Med. 2003;9:1195–1201. Sasaki K, Heeschen C, Aicher A, Ziebart T, Honold J, Urbich C, Rossig L, Koehl U, Koyanagi M, Mohamed A, Brandes RP, Martin H, Zeiher AM, Dimmeler S. Ex vivo pretreatment of bone marrow mononuclear cells with endothelial NO synthase enhancer AVE9488 enhances their functional activity for cell therapy. Proc Natl Acad Sci U S A. 2006; 103:14537–14541. Thum T, Fleissner F, Klink I, Tsikas D, Jakob M, Bauersachs J, Stichtenoth DO. Growth hormone treatment improves markers of systemic nitric oxide bioavailability via insulin-like growth factor-I. J Clin Endocrinol Metab. 2007;92:4172– 4179. Walter DH, Rochwalsky U, Reinhold J, Seeger F, Aicher A, Urbich C, Spyridopoulos I, Chun J, Brinkmann V, Keul P, Levkau B, Zeiher AM, Dimmeler S, Haendeler J. Sphingosine-1-phosphate stimulates the functional capacity of progenitor cells by activation of the CXCR4dependent signaling pathway via the S1P3 receptor. Arterioscler Thromb Vasc Biol. 2007;27:275–282. Seeger FH, Haendeler J, Walter DH, Rochwalsky U, Reinhold J, Urbich C, Rossig L, Corbaz A, Chvatchko Y, Zeiher AM, Dimmeler S. p38 mitogen-activated protein kinase downregulates endothelial progenitor cells. Circulation. 2005;111:1184 –1191. Sorrentino SA, Bahlmann FH, Besler C, Muller M, Schulz S, Kirchhoff N, Doerries C, Horvath T, Limbourg A, Limbourg F, Fliser D, Haller H, Drexler H, Landmesser U. Oxidant stress impairs in vivo reendothelialization capacity of endothelial progenitor cells from patients with type 2 diabetes mellitus: restoration by the peroxisome proliferator-activated receptor-gamma agonist rosiglitazone. Circulation. 2007;116:163–173. Jiao C, Fricker S, Schatteman GC. The chemokine (C-X-C motif) receptor 4 inhibitor AMD3100 accelerates blood flow restoration in diabetic mice. Diabetologia. 2006;49:2786 –2789. Liles WC, Broxmeyer HE, Rodger E, Wood B, Hubel K, Cooper S, Hangoc G, Bridger GJ, Henson GW, Calandra G, Dale DC. Mobilization of hematopoietic progenitor cells in healthy volunteers by AMD3100, a CXCR4 antagonist. Blood. 2003;102:2728 –2730. Broxmeyer HE, Orschell CM, Clapp DW, Hangoc G, Cooper S, Plett PA, Liles WC, Li X, Graham-Evans B, Campbell TB, Calandra G, Bridger G, Dale DC, Srour EF. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med. 2005;201:1307–1318. Shepherd RM, Capoccia BJ, Devine SM, Dipersio J, Trinkaus KM, Ingram D, Link DC. Angiogenic cells can be rapidly mobilized and efficiently harvested from the blood following treatment with AMD3100. Blood. 2006;108:3662–3667. Tan Y, Li Y, Xiao J, Shao H, Ding C, Arteel GE, Webster KA, Yan J, Yu H, Cai L, Li X. A novel CXCR4 antagonist derived from human SDF-1beta enhances angiogenesis in ischaemic mice. Cardiovasc Res. 2009;82:513–521. Tendera M, Wojakowski W, Ruzyllo W, Chojnowska L, Kepka C, Tracz W, Musialek P, Piwowarska W, Nessler J, Buszman P, Grajek S, Breborowicz P, Majka M, Ratajczak MZ. Intracoronary infusion of bone marrow-derived selected CD34⫹CXCR4⫹ cells and non-selected mononuclear cells in patients with acute STEMI and reduced left ventricular ejection fraction: results of randomized, multicentre Myocardial Regeneration by Intracoronary Infusion of Selected Population of Stem Cells in Acute Myocardial Infarction (REGENT) Trial. Eur Heart J. 2009;30:1313–1321. Losordo DW, Schatz RA, White CJ, Udelson JE, Veereshwarayya V, Durgin M, Poh KK, Weinstein R, Kearney M, Chaudhry M, Burg A, Eaton L, Heyd L, Thorne T, Shturman L, Hoffmeister P, Story K, Zak V, Dowling D, Traverse JH, Olson RE, Flanagan J, Sodano D, Murayama T, Kawamoto A, Kusano KF, Wollins J, Welt F, Shah P, Soukas P, Asahara T, Henry TD. Intramyocardial transplantation of autologous CD34⫹ stem cells for intractable angina: a phase I/IIa double-blind, randomized controlled trial. Circulation. 2007;115: 3165–3172.

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

Jarajapu and Grant 160. Losordo DW, Henry T, Schatz RA, Lee JS, Costa M, Bass T, Schaer G, Niederman A, Mendelsohn F, Davidson C, Waksman R, Soukas PA, Simon D, Chronos N, Fortuin FD, Huang PP, Weintraub N, Yeung A, Rosenfield K, Wong SC, Taussig A, Rava AN, Sherman W, Kereiakes D, Strumpf RK, Port S, Pieper K, Adams PX, Harrington R. Autologous CD34⫹ cell therapy for refractory angina: 12 Month results of the Phase II ACT34-CMI study. Circulation. 2009;120:S1132. 161. Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, Homma S, Edwards NM, Itescu S. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med. 2001;7:430 – 436. 162. Jackson KA, Majka SM, Wang H, Pocius J, Hartley CJ, Majesky MW, Entman ML, Michael LH, Hirschi KK, Goodell MA. Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells. J Clin Invest. 2001;107:1395–1402. 163. Botta R, Gao E, Stassi G, Bonci D, Pelosi E, Zwas D, Patti M, Colonna L, Baiocchi M, Coppola S, Ma X, Condorelli G, Peschle C. Heart infarct in NOD-SCID mice: therapeutic vasculogenesis by transplantation of human CD34⫹ cells and low dose CD34⫹KDR⫹ cells. FASEB J. 2004;18:1392–1394. 164. Ott I, Keller U, Knoedler M, Gotze KS, Doss K, Fischer P, Urlbauer K, Debus G, von Bubnoff N, Rudelius M, Schomig A, Peschel C, Oost-

Cell-Based Therapies for Diabetic Complications

165.

166.

167.

168.

169.

869

endorp RA. Endothelial-like cells expanded from CD34⫹ blood cells improve left ventricular function after experimental myocardial infarction. FASEB J. 2005;19:992–994. Gulati R, Jevremovic D, Peterson TE, Witt TA, Kleppe LS, Mueske CS, Lerman A, Vile RG, Simari RD. Autologous culture-modified mononuclear cells confer vascular protection after arterial injury. Circulation. 2003;108:1520 –1526. Yoon YS, Wecker A, Heyd L, Park JS, Tkebuchava T, Kusano K, Hanley A, Scadova H, Qin G, Cha DH, Johnson KL, Aikawa R, Asahara T, Losordo DW. Clonally expanded novel multipotent stem cells from human bone marrow regenerate myocardium after myocardial infarction. J Clin Invest. 2005;115:326 –338. Kalka C, Masuda H, Takahashi T, Kalka-Moll WM, Silver M, Kearney M, Li T, Isner JM, Asahara T. Transplantation of ex vivo expanded endothelial progenitor cells for therapeutic neovascularization. Proc Natl Acad Sci U S A. 2000;97:3422–3427. Urbich C, Heeschen C, Aicher A, Dernbach E, Zeiher AM, Dimmeler S. Relevance of monocytic features for neovascularization capacity of circulating endothelial progenitor cells. Circulation. 2003;108: 2511–2516. Murohara T, Ikeda H, Duan J, Shintani S, Sasaki K, Eguchi H, Onitsuka I, Matsui K, Imaizumi T. Transplanted cord blood-derived endothelial precursor cells augment postnatal neovascularization. J Clin Invest. 2000;105:1527–1536.

Downloaded from circres.ahajournals.org at Univ Florida on March 19, 2010

Lihat lebih banyak...

Comentários

Copyright © 2017 DADOSPDF Inc.