Skip to main content
American Journal of Physiology - Heart and Circulatory Physiology logoLink to American Journal of Physiology - Heart and Circulatory Physiology
. 2015 Dec 18;310(4):H455–H465. doi: 10.1152/ajpheart.00726.2015

Stem cell-based therapies to promote angiogenesis in ischemic cardiovascular disease

Luqia Hou 1,2, Joseph J Kim 1,2, Y Joseph Woo 2,3, Ngan F Huang 1,2,3,
PMCID: PMC4796616  PMID: 26683902

Abstract

Stem cell therapy is a promising approach for the treatment of tissue ischemia associated with myocardial infarction and peripheral arterial disease. Stem and progenitor cells derived from bone marrow or from pluripotent stem cells have shown therapeutic benefit in boosting angiogenesis as well as restoring tissue function. Notably, adult stem and progenitor cells including mononuclear cells, endothelial progenitor cells, and mesenchymal stem cells have progressed into clinical trials and have shown positive benefits. In this review, we overview the major classes of stem and progenitor cells, including pluripotent stem cells, and summarize the state of the art in applying these cell types for treating myocardial infarction and peripheral arterial disease.

Keywords: stem cell therapy, pluripotent stem cell, angiogenesis, myocardial infarction, peripheral arterial disease, hindlimb ischemia


cardiovascular disease (CVD) is the leading cause of mortality and morbidity among Americans and Veterans, accounting for one in every three deaths and a hospital burden of $196 billion (63). In particular, tissue ischemia associated with coronary heart disease and peripheral arterial disease (PAD) accounts for more than half of all CVDs. Although the quality of life of the patients with CVDs has improved over recent decades, owing to drug treatment and organ transplantation, there is still a tremendous need to develop improved therapies for the treatment of tissue ischemia. Accordingly, stem cell-based approaches to promote angiogenesis for the improvement of tissue function and/or blood perfusion are attractive. A number of stem and progenitor cells have been examined for boosting angiogenesis within ischemic CVDs, including adult bone marrow-derived mononuclear cells (BM-MNCs), endothelial progenitor cells (EPCs), and pluripotent stem cell-derived endothelial cells (PSC-ECs). In this review, we will discuss the state of stem cell therapy for the treatment of peripheral arterial disease (PAD) and myocardial infarction (MI) in the preclinical and clinical settings.

Classes of Stem and Progenitor Cells

To describe the therapeutic efficacy of stem cells in preclinical models, here we first define and characterize these various cell populations. Stem and progenitor cells are generally identified by specific phenotypic markers, although no single marker is sufficient to uniquely distinguish them from other cells. A phenotypic signature can be empirically defined based on the expression of surface receptors, transcription factors, and cytosolic proteins, in addition to morphological or functional characteristics. Below is an overview of the major classes of stem and progenitor cells used in the treatment of CVDs (Table 1).

Table 1.

Characterization of therapeutic stem cell types

Type Source Differentiation Capacity PAD Applications MI Applications
EPC Bone marrow/peripheral blood Multipotent (2, 41, 47, 88, 119) (43, 68)
MSC Bone marrow/adipose tissue/blood Multipotent (25, 45, 46) (56, 61, 87)
ESC Inner cell mass of blastocyst Pluripotent (13, 35, 117) (32, 50, 57, 116)
iPSC Any somatic cell type Pluripotent (83) (24, 96)

PAD, peripheral arterial disease; MI, myocardial infarction; EPC, endothelial progenitor cell; MSC, mesenchymal stem cell; ESC, embryonic stem cell; iPSC, induced pluripotent stem cell.

Mononuclear cells.

BM-MNCs and peripheral blood MNCs (PB-MNCs) are the most commonly used therapeutic cells for clinical treatment of CVDs, owing to their ease of isolation by bone marrow aspiration or venipuncture and subsequent purification by density gradient centrifugation (23). Another advantage is that these cells can be harvested with short turnaround time and implanted into the patient without in vitro expansion. BM-MNCs and PB-MNCs represent heterogeneous populations composed of hematopoietic stem cells, mesenchymal stem cells (MSCs), and EPCs. As a result, they are primarily characterized by having a single nucleus. The cellular composition appears to vary depending on the purification procedure. Comparison of MNC isolation approaches demonstrated that Ficoll gradient centrifugation separation resulted in higher cell recovery than using Lymphoprep separation (85). Furthermore, Lymphoprep separation resulted in a reduction in CD34+ and CD133+ hematopoietic stem cells, compared with the use of Ficoll gradient centrifugation. These results underscore the importance of the purification approach on cellular composition and demonstrate the potential advantages of Ficoll gradient centrifugation for the isolation of MNCs.

Endothelial progenitor cells.

First described by Asahara and colleagues (2), EPCs derived from the bone marrow or PB are known to home to sites of tissue injury to regenerate new blood vessels. Representing less than 1% of the cell population in PB, EPCs can be purified from blood by density gradient centrifugation for MNCs, followed by attachment onto tissue culture plastic dishes (104). Although there is no consensus in the phenotypic marker expression of EPCs, they are generally characterized by the positive expression of FLK1 (VEGF receptor 2), CD133, and CD34 at early stages after attachment onto tissue culture plastic dishes (3, 34, 78). After expansion for several weeks, the late-outgrowth EPCs functionally express endothelial markers such as vascular endothelial (VE) -cadherin and von Willebrand factor (vWF). EPCs also express C-X-C chemokine receptor type 4 receptors that bind stromal-derived factor-1, which is released at sites of tissue ischemia (11). EPCs home to these sites of tissue injury and participate by directly forming new vessels and indirectly by secreting proangiogenic cytokines and growth factors (4).

Mesenchymal stem cells.

MSCs are a class of stromal cells with spindle-shaped morphology (10, 42, 73, 75). They can be derived from numerous organs including bone marrow, liver, adipose tissue, blood, and liver (7, 9). MSCs within bone marrow occupy only 0.01% of the total population but can expand over one million-fold (73). As a multipotent stem cell, MSCs have limited self-renewal capacity and can give rise to specialized cells such as adipocytes, chondrocytes, and osteocytes (73, 74). From bone marrow aspirates, MSCs can be isolated by density gradient centrifugation techniques, followed by plating onto tissue culture plastic for adherent cells. Although MSC phenotype varies within different organs and species, they generally express the surface markers STRO-1 (a stromal cell surface antigen), CD29 (integrin-β1), CD44 (receptor for hyaluronic acid and matrix proteins), CD105 (endoglin), and CD166 (cell adhesion molecule) (18, 89). MSCs predominantly give rise to cells of musculoskeletal and fat lineages but have been shown to differentiate into endothelial lineage by stimulation with growth factors (110), shear stress (106), and extracellular matrix compliance (109, 110). Besides differentiating into cells of the endothelial lineage, MSCs can also participate in angiogenesis by secreting angiogenic paracrine factors including VEGF, basic fibroblast growth factor (bFGF), and PDGF (22, 45).

Pluripotent stem cells.

Human embryonic stem cells (ESCs) are PSCs derived from the inner cell mass of the blastocyst (97). They are capable of infinite expansion, while being capable of giving rise to cells from all three germ layers. Morphologically, ESCs grow in compact colonies formed on top of murine embryonic fibroblast feeder cells or on Matrigel basement membrane extract (114). Phenotypically, ESCs express pluripotent markers such as transcriptional factors Oct3/4 and Sox2, homeodomain protein Nanog, and surface markers SSEA-1 and TRA-1-81 (28, 79, 111). Limiting the translational relevance of ESCs are the ethical concerns of embryo destruction, along with the potential dangers of teratoma formation.

An alternative PSC source are iPSCs, which are somatic cells that have been reprogrammed into a PSC state by the genetic activation of key transcription factors (i.e.., Oct3/4, Sox2, Klf4, and c-Myc) (91, 92). Human iPSCs appear to resemble many pluripotent properties of ESCs in their ability to have nearly unlimited self-renewal capacity and differentiate into cells of all three germ layers, including endothelial lineage (92). In contrast to ESCs, however, iPSCs can be derived from a patient's somatic cells, which overcomes concerns of immune rejection and the destruction of embryos. In the advent of technological advances, iPSCs can now be generated using nongenetic-based approaches, which will improve the safety of these cells for clinical use (1, 52, 80). Thus iPSCs may be the most promising patient-specific cell source for regenerative medicine.

To obviate the potential formation of teratomas, ESCs and iPSCs can be terminally differentiated into endothelial lineage before in vivo implantation. A number of robust differentiation protocols have been described that take advantage of chemical factors such as VEGF, bone morphogenetic protein-4, bFGF, and/or Wnt signaling pathways for inducing endothelial differentiation (51, 83, 108, 118).

In summary, a number of stem cells can be delivered for the treatment of CVDs, although the identity and characterization of the stem cell populations may vary depending on the phenotypic markers used for characterization. MNCs, EPCs, and MSCs constitute a readily accessible population of adult autologous therapeutic cells. However, concerns of limited plasticity in differentiation, along with reduced function and number of adult stem cells in cardiovascular patients, dampen the enthusiasm for their usage (8, 98, 104). Human iPSCs represent a new class of autologous stem cells with greater plasticity in differentiation, but the concerns of teratoma formation currently limit their translation into clinical practice.

Mechanism of Stem Cell Participation

To repair tissue ischemia, stem cells participate in angiogenesis by releasing angiogenic factors and/or by differentiating into vascular lineage. For example, MSCs secrete angiogenic paracrine factors such as VEGF, bFGF, and PDGF that stimulate neovessel formation (22, 45). Some investigators also reported the ability of MSCs to differentiate into endothelial lineage and/or express endothelial lineage markers (36, 49, 69, 115). We have previously shown that ECs derived from human iPSCs (iPSC-ECs) secrete angiogenic factors to similar degrees as primary ECs, including bFGF, VEGF, and angiopoietin-1 (83). In addition, the iPSC-ECs can functionally incorporate into the perivascular space. EPCs and MNCs can differentiate into endothelial lineage by forming neovasculature that functionally anastamose with host circulation (3, 12). These varying mechanisms enable stem cells to differentiate and/or to incorporate into vasculature or release paracrine factors to stimulate host-derived angiogenesis.

Below, we discuss the applications of these classes of stem cells in treating PAD and MI in preclinical and clinical settings.

Peripheral Arterial Disease

PAD, which affects over eight million patients in the United States, is an occlusive atherosclerotic disease caused by blockage of the arteries from cholesterol plaques (30, 31, 64). PAD limits blood supply to the lower extremities. Symptoms of PAD include intermittent claudication, which is defined as calf or buttock pain, or cramping with walking caused by inadequate blood flow to the limb (70). The most severe symptomatic manifestation of PAD is critical limb ischemia (CLI), defined as pain at rest due to reduced blood flow to the limb. CLI may further result in ischemic ulceration and gangrene formation. Patients with CLI have not only a high risk of amputation but a high rate of cardiovascular death, often due to complications related to coronary artery (55%) and cerebrovascular atherosclerosis (10%) (19).

Among preclinical models of PAD, the most popular and widely used one is the hindlimb ischemia model (HLI). In this experimental model, the femoral artery is ligated and excised, reducing blood flow to the lower leg by ∼20% of normal blood perfusion, based on laser-Doppler spectroscopy (67). The HLI model has been widely used to evaluate the functional capacity of therapeutic stem cells (Table 2).

Table 2.

Application of stem cells for treatment of PAD

Disease Model Cell Type Injection Approach Follow-up Time Output Measurement Reference
Mouse/rabbit HLI Putative progenitor ECs Tail vein injection 1 to 6 wk Integration to capillary vessel walls, arranged into capillaries by 6 wk; incorporation of cells in capillaries and small arteries; cells localized to neovascular zones (2)
Rat HLI CD133+ progenitor cells Injectable collagen-based matrix 2 wk CD133+ cells retained better; cells incorporated into vascular structures' increased intramuscular arteriole and capillary density (90)
Rabbit HLI BM-MNCs Intramuscular injection into ischemic thigh skeletal muscle 4 wk More detectable collateral vessels, higher capillary density, greater laser-Doppler blood perfusion index (88)
Mouse HLI BM-MNCs or PB-MNCs Intramuscular injection into ischemic skeletal muscles 4 wk Increased blood flow ratio and increased capillary density (119)
Mouse HLI MSC Intramuscular injection into adductor muscle at 24, 48, and 72 h post-femoral ligation 4 wk Improve limb function, reduce autoamputation, and attenuate muscle atrophy and fibrosis (45)
Rat HLI MSC Intramuscular injection into ischemic thigh muscle 3 wk Higher laser-Doppler perfusion index, improvement in blood perfusion, and higher capillary density (40)
Mouse HLI ESC-ECs Intramuscular injection into ischemic limbs 4 wk Increased limb salvage; increased blood perfusion; increased capillary and arteriole densities (13)
Mouse HLI ESC-ECs and ESC-SMC Intrafemoral artery 1 to 6 wk Increased blood flow and capillary density. Cells incorporated into host circulating vessels (117)
Mouse HLI ESC; ESC-ECs Intramuscular, intrafemoral artery, intrafemoral vein injections 2 wk ESC-ECs localized to the ischemic limb; engraftment of ESC-ECs into vasculature; improved limb perfusion and neovascularization (37)
Mouse HLI iPSC-ECs Intramuscular injection at day 0 and day 7 2 wk Increased blood flow and capillaries (83)

ECs, endothelial cells; HLI, hindlimb ischemia; BM, bone marrow; PB, peripheral blood; MNCs, mononuclear cells.

Preclinical Studies to Treat PAD Using Therapeutic Stem Cells

Treatment of PAD using MNCs and EPCs.

Asahara and colleagues (2) first demonstrated that PB-derived CD34+ EPCs integrated with host capillary vessel walls and formed capillaries in the ischemic limbs of syngenic mice and autologous rabbits. Shintani et al. (88) provided evidence that BM-MNCs derived from Histopaque density gradient centrifugation augmented neovascularization in a rabbit HLI model by inducing collateral vessel formation and blood perfusion. Kobayashi et al. (47) used a similar rabbit HLI model and studied the effect of Ang-1 gene transfer combined with transplantation of low dose BM-MNCs isolated using Histopaque density gradient centrifugation. In this study, the authors observed notable improvement in angiographic score, an increase in capillary density, an increase in transcutaneous oxygen pressure, and a reduction in skin ulceration. The angiographic score was a quantitative analysis of collateral vessel development from angiogram films (93). It is often calculated as the ratio of grid intersections crossed by contrast-opacified arteries to the total number of grid intersections in the medial thigh area. In addition, Jeon et al. (41) studied the effect of combined angiogenic therapies in a mouse HLI model by performing BM-MNC transplantation along with EPC mobilization with granulocyte colony-stimulating factor. They reported an increase in microvessel density and more extensive expression of bFGF and VEGF when the combined therapy was performed, compared with administration of each treatment separately. Similar improvements in capillary density and blood perfusion were reported by Zhang et al. (119) when Histopaque gradient-isolated BM-MNCs were transplanted in mice with ischemic skeletal muscle.

Treatment of PAD using MSCs.

As for MSCs, a number of studies have been performed to show their therapeutic effects. Kinnaird et al. (46) showed that CD44+/CD90+ MSCs were able to improve limb function, reduce autoamputation, and attenuate muscle atrophy and fibrosis in a murine HLI model. The authors (45) demonstrated that the therapeutic benefits were due to paracrine mechanisms, especially through the release of bFGF and VEGF from the cells. In a related study, the same group also provided a full spectrum of cytokine genes expressed by MSCs to examine the paracrine mechanisms. Importantly, they demonstrated that injection of cells was not required for therapeutic benefits. Instead, treatment with conditioned media from MSCs was sufficient to mediate arteriogenesis and enhance collateral flow, which provides supportive evidence of the paracrine effects of MSCs. Iwase et al. (40) reported in a rat model that delivery of adherent spindle-shaped MSCs from bone marrow produced significantly improved blood perfusion, compared with Ficoll gradient isolated BM-MNCs. The authors also compared the angiogenic potency of MSCs and BM-MNCs under an ischemic environment in vivo. MSCs appeared to have more therapeutic effect than MNCs and also differentiate into ECs and vascular smooth muscle cells by detecting vWF and smooth muscle α-actin, respectively. More recently, Han et al. (25) reported that hypoxia-preconditioned human umbilical cord-derived CD44+/CD105+ MSCs were able to induce angiogenesis and low-inflammatory immune response in immunodeficient mice with HLI, suggesting that hypoxia preconditioning may be able to boost the therapeutic effects of MSCs.

Treatment of PAD using PSC-derived ECs.

ECs differentiated from human ESCs (ESC-ECs) and iPSCs (iPSC-ECs) have also been tested in HLI models. Cho et al. (13) tested the intramuscular injection of human ESC-ECs in a mouse HLI model. These cells were characteristic of primary ECs in the gene and protein expression of CD31, VE-cadherin, Tie-2, Flk-1, and vWF. When the cells were delivered to the ischemic limb, the animals demonstrated improved limb salvage and blood perfusion, along with increased capillary and arteriole densities. Since mature vessels are composed not only of ECs but also smooth muscle cells, Yamahara et al. (117) transplanted both VE-cadherin+ ESC-ECs with smooth muscle α-actin+ ESC-derived smooth muscle cells together into the ischemic limb. When delivered together, there were reportedly synergistic effects in neovascularization and blood perfusion, compared with when only one cell type was delivered. To track the kinetics of cell survival, Huang et al. (37) used bioluminescence imaging to track VE-cadherin+ murine ESC-ECs delivered to the ischemic hindlimb by intramuscular, intrafemoral artery, or intrafemoral vein injection. The results showed that ESC-ECs delivered by all three modalities localized in ischemic limb, but the systemically delivered cells resulted in greater improvement in limb perfusion and neovascularization, compared with intramuscular delivery. Rufaihah et al. (83) examined the therapeutic efficacy of CD31+ human iPSC-ECs in mouse HLI model. When compared with that in the saline or control fibroblast groups, intramuscular injection of human iPSC-ECs into severe combined immunodeficient mice demonstrated a significant increase in capillary density and blood perfusion for at least 28 days. These data further support the feasibility of using human iPSC-ECs in developing novel cell therapies for patients with PAD.

Myocardial Infarction

Another ischemic CVD is MI, which affects more than seven million people in the United States alone (64, 81). MI results from obstruction of the coronary arteries, leading to prolonged ischemia and death of cardiomyocytes. The massive loss of cardiomyocytes, vascular cells, and interstitial cells (in the order of 1 billion) contributes to the frequent hospitalizations and premature death of patients undergoing traditional therapies. It is widely agreed that the human myocardium has low regenerative capacity that associates with inadequate compensation for severe loss of heart muscle (44). Therefore, regenerative approaches to restore myocardium are in urgent need and have attracted a large amount of preclinical and clinical testing.

Preclinical MI models are commonly generated from occlusion of the left anterior descending coronary artery, followed by reperfusion (60). MI models can be generated from small rodents to large pigs and sheep. To repair the heart, early studies transplanted fetal, neonatal, and adult cardiomyocytes and showed stable grafts in the injured hearts (65, 77). However, because of their limited availability, researchers turned to stem cells as alternative sources to improve cardiac function (16, 20, 21, 66, 86, 101, 102). The main approaches to deliver the cells include transepicardial injection into the myocardium (58, 59), transendocardial injection using percutaneous catheters (27, 29), and intracoronary infusion using angioplasty balloons (5, 55).

Treatment of MI using MNCs.

Adult stem cells have been extensively tested in the preclinical setting (Table 3). For basic research and clinical trials, the major source of adult stem cells is the bone marrow. In small and large animal models, BM-MNCs have been shown to be able to reduce infarct size and improve left ventricular function and perfusion, along with modest improvements in physiological and anatomical parameters in human patients such as peak systolic displacement, peak systolic strain, as well as left ventricular ejection fraction (LVEF) (82). Orlic et al. (68) provided strong evidence that lineage-negative (Lin) c-kit+ bone marrow cells were able to generate de novo myocardium, occupying 68% of the infarcted portion of the ventricle 9 days after local transplantation in mice. New endothelial and smooth muscle cells were also identified in coronary vessels. Kamihata et al. (43) pioneered the transplantation of CD34+ BM-MNCs into the ischemic myocardium in swine. Regional blood flow, capillary densities were significantly increased, with improved cardiac function. The data also revealed that BM-MNCs were incorporated into more than 30% of neocapillaries and 8% of macrophages. Angiogenic ligands and cytokines, such as cardiac interleukin-1β and tumor necrosis factor-α, were identified and believed to also play a role in angiogenesis.

Table 3.

Application of stem cells for treatment of MI

Disease Model Cell Type Injection Approach Follow-up Time Output Measurement Reference
Mouse MI (Lin) c-kit+ BM cells Intramyocardial injection in the anterior and posterior aspects of the viable myocardium bordering the infarct 9 days Generated de novo myocardium, comprised proliferating myocytes and vascular structure (68)
Porcine MI BM-MNCs Transendocardial injection 4 wk Increased ejection fraction, increased regional blood flow and reduction in ischemic area (43)
Porcine MI MSCs Intramyocardial injection 2–4 wk Robust engraftment of MSCs, attenuated degree of contractile dysfunction, and reduced wall thinning after MI (87)
Rat MI MSCs (Akt1) Intramyocardial injection Inhibit cardiac remodeling by reducing intramyocardial inflammation, collagen deposition and cardiac myocyte hypertrophy, regenerated 80–90% lot myocardium, and normalized systolic and diastolic cardiac function (56)
Rat MI MSCs Monolayered cell grafts placed on surface of the anterior scar 8 wk MSC sheet grew to form a thick stratum with vessels, undifferentiated cells and cardiomyocytes, reversed wall thinning and improved cardiac function (61)
Mouse MI ESC-ECs Intramyocardial injection 8 wk Echocardiogram show improved LV function; increased capillaries and venules in infarcted zones (50)
Mouse MI iPSC-ECs Intramyocardial injection 4 wk Echocardiogram show improved LV function; MRI show increased ejection fraction; proangiogenic and antiapoptotic factors released from iPSC-ECs (24)
Porcine MI ESC-ECs and SMC Fibrin 3-dimensional porous scaffold biomatrix with hESC-ECs and SMCs seeded injected 1 to 4 wk MRI show improved LV function; significant engraftment of hESC-ECs (113)
Porcine MI iPSCs Intramyocardial injection 15 wk iPSCs can be visualized ≤15 wk; hiPSC-ECs contribute to vascularization (96)

SMC, smooth muscle cell; h, human; LV, left ventricular.

Treatment of MI using MSCs.

As for MSCs, a number of rodent and swine models have been used to prove the ability of MSCs to engraft and differentiate in the heart. Shake et al. (87) injected autologous porcine Di-I-labeled MSCs expanded from bone marrow aspirates of swine into post-MI swine myocardium and observed successful engraftment of these cells in the scarred myocardium, as well as the expression of cardiomyocyte markers such as α-actin, tropomyosin, troponin T, myosin heavy chain, and phospholamban at 2 wk postinjection. Mangi et al. (56) genetically engineered rat MSCs by overexpressing the prosurvival gene Akt1. They found that transplantation of these cells into the ischemic rat myocardium repaired infarcted myocardium by restoring fourfold greater myocardial volume than cells transduced with reporter gene lacZ. Miyahara et al. (61) transplanted a cell sheet composed of CD29+/CD90+ MSCs onto the infarcted rat epicardium and reported that the engrafted sheet augmented neovascularization as a result of paracrine signaling, while also reversing wall thinning and improving cardiac function.

Treatment of MI using PSC-derived ECs.

For decades, ESCs have held great promise for myocardial cell therapy, despite the fact that they have raised ethical and political debates for clinical application. Li et al. (50) showed that syngenic VE-cadherin+ ESC-ECs injected into the peri-infarct zone of mouse hearts reported a higher density of capillaries in the infarcted area, along with functional improvement in fractional shortening. A number of other preclinical studies also demonstrate that ESC transplantation had beneficial effects in cardiac structure and function (32, 57, 116). Recently, Chong et al. (14) successfully performed transepicardial intramuscular injection in primates using human ESC-derived cardiomyocytes from H7 or RUES2 human ESC lines. Although a preliminary study, it showed not only a large scale remuscularization but also electromechanical integration with the host tissue.

The creation of human iPSCs successfully obviates the ethical problems associated with ESC research. Gu and colleagues (24) generated iPSC-ECs from porcine iPSCs that were then transplanted into a mouse model of MI. This transplantation of CD31+ iPSC-ECs significantly improved fractional shortening by up to 5% and LVEF by 3.5%. A more fundamental finding of this study was achieved by using quantitative protein assays and single-cell PCR. The authors identified multiple proangiogenic and antiapoptotic factors released from iPSC-ECs that were responsible for the neovascularization and the survival of cardiomyocytes in the ischemic region. Recently, large animal models of MI were also used to test similar transplantation procedures. Templin et al. (96) evaluated the transplantation of human iPSCs (96) using in vivo molecular imaging technique to track these transgenic human iPSC lines for up to 15 wk. Neovascularization was reported in the animals with iPSC-ECs. However, one common uncertainty from the above studies is that the authors were not able to determine if the improvements in cardiac function were the result of the differentiation of stem cells into cardiomyocytes or a result of paracrine factors, such as interleukin 6 and IGF-1, secreted by the transplanted cells.

Together, these data demonstrate the feasibility and efficacy of delivering stem and progenitors to improve the function of the ischemic myocardium or limb in preclinical animal models. A number of limitations in these studies include the lack of functional immune response associated with the use of immunodeficient animal strains or immunosuppression in cases where human cells were transplanted. Furthermore, longer-term follow-up from animal studies will be required to demonstrate long-term efficacy and safety. Nevertheless, these studies provide promising results, suggesting the potential benefits of stem cell therapy for treatment of MI and PAD.

Clinical Translation of Stem Cell Therapy

Stem cell therapy for clinical treatment of PAD.

It has been two decades since the first clinical trial of stem cell therapy took place to treat patients with PAD. So far, more than 50 clinical trials have been reported to use progenitor cells for the treatment of PAD with varying severity (15). The majority of clinical trials have focused on CLI patients in small phase I or II studies. The cell types that have been selected for clinical trials include BM-MNCs, PB-MNCs, marker-specific subsets of cells derived from bone marrow or blood, as well as MSCs. To date there is still no Food and Drug Administration-approved stem cell therapies for patients with PAD. However, we have learned a tremendous amount of knowledge about the mechanisms of angiogenesis and vascular regeneration using cell therapy. Since stem cell clinical trials in PAD patients have been described elsewhere (17, 76), below we will briefly describe several of such clinical trials.

Therapeutic angiogenesis using cell transplantation (TACT), a randomized controlled trial, was the first-reported autologous stem cell therapy clinical trial using BM-MNCs and PB-MNCs (94). At 6-mo follow-up, patients treated with BM-MNCs showed improvement in rest pain, transcutaneous oxygen pressure, ankle brachial index (ABI), and pain-free walking distance. Similarly, Van Tongeren et al. (103) have tested delivery of CD34+ BM-MNCs in a small group of patients and revealed significant improvements in ABI, pain score, and pain-free walking distance after 12 mo. In a multicenter, double-blind, randomized start clinical trial, Intraarterial Progenitor Cell Transplantation of Bone Marrow Mononuclear Cells for Induction of Neovascularization in Patients With Peripheral Arterial Occlusive Disease (PROVASA), patients that received intraarterial delivery of BM-MNCs showed improved ulcer healing and rest pain, but no significant change in ABI or limb salvage (105). In another multicenter, randomized, double-blind trial, bone marrow aspirate concentrate (BMAC) clinical trial, improvement in amputation, pain, and quality of life were reported at 3-mo and 6-mo follow-ups (39). Together, these studies suggested that intramuscular administration of BM-MNCs results in improved ABI and lower amputation rates. In addition to BM-MNCs, PB-MNCs were also tested in a number of clinical trials through intramuscular administration in patients with PAD. In a pilot clinical trial, Huang et al. (38) reported that administration of 108 PB-MNC with 0.4% CD34+ cells were able to improve ABI, ulcer healing, blood perfusion, angiographic scores, and limb salvage at 3-mo follow-up (38). Similar improvements in ABIs, pain scores, and amputation rates were reported in other phase I clinical trials at 3-mo follow-up (62, 71).

MSCs were also used in multiple clinical trials due to their multipotent potential and immunosuppressive effects. Significant improvement in pain-free walking time was reported in a double-blind, randomized controlled trial comparing BM-MSCs and BM-MNCs after a 6-mo follow-up (54). Not only did treatment by BM-MSCs result in greater collateral blood vessel formation, but it also showed faster ulcer healing than the group treated with BM-MNCs. In addition to the above cell types, other cells such as allogenic MSCs, adipose-derived regenerative cells, ESCs/iPSCs, or nuclear reprogrammed stem cells are being currently explored in preclinical models and will head to clinical trials if they show advantages to other adult stem cells.

Stem cell therapy for clinical treatment of MI.

The treatment of myocardial injury has been increasingly strategized using stem cell approaches. Over 100 clinical trials involving stem cell therapy for the treatment of MI have been reported (33). Depending on the stage of injury, stem cell therapies have sought to prevent the progression of acute MI toward ischemic cardiomyopathy and congestive heart failure, or restore function in failing or chronically ischemic hearts. Since stem cell clinical trials in MI patients have been described elsewhere (95), below we will briefly describe several selected clinical trials.

The results of recent United States clinical trials performed by the Cardiovascular Cell Therapy Research Network indicate the lack of therapeutic efficacy of BM-MNCs for treatment of acute MI (100). Briefly, 120 patients were randomly assigned to either BM-MNC therapy or placebo at different times of introduction (i.e., 3 or 7 days after percutaneous coronary intervention). After 6 mo, MRI-based evaluation of LVEF revealed that those treated with BM-MNCs did not show significant improvement compared with the placebo group. Thus, because of the lack of promise of BM-MNCs from several clinical trials (72, 84, 99, 112), a shift in focus has been made toward other stem cell sources in the past several years.

MSCs have also been tested in clinical trials, owing to their potentially immunomodulatory property. In the Prevention of Contrast Renal Injury with Different Hydration Strategies (POSEIDON) trial, autologous and allogeneic human MSCs were injected in patients with left ventricular dysfunction due to ischemic cardiomyopathy. The MSCs were administered in three different doses and monitored over 1 year to assess the efficacy and safety of autologous MSC therapy based on various output measures, including a 6-min walk test, exercise peak, oxygen consumption, and LVEF. The conclusion of these trials was that MSC injection favorably affected patient functional capacity and ventricular remodeling, regardless of being allogeneic or autologous (26).

Another highly investigated cell source for the treatment of MI are CD34+ EPCs. In a randomized, double-blind, phase-II study, 167 patients with refractory angina were injected with autologous CD34+ cells versus a placebo (53). The cells were targeted to 10 sites of ischemic, viable myocardium, and the primary outcome measure was weekly angina frequency 6 mo following treatment. The results of this clinical trial demonstrated that CD34+ cell administration contributed to a reduction of angina frequency, with an improvement in exercise tolerance. Together, these clinical studies suggest that some adult stem cells may be promising candidates for treating the ischemic myocardium.

Approaches to Enhance Stem Cell Therapeutic Potential

Paracrine factors play an important role in improving tissue repair and functional recovery after stem cell injection. Therefore, a number of groups have developed approaches to enhance cell survival and function, along with paracrine factor delivery. Gnecchi et al. (22) showed that conditional media from hypoxic Akt-expressing MSCs significantly inhibited hypoxia-induced apoptosis and triggered vigorous contraction in adult rat cardiomyocytes in vitro. When injected into infarcted heart, the conditioned media limited the infarct size and improved ventricular function. Further characterization of the conditioned media revealed that a number of genes encoding paracrine factors were upregulated, including VEGF, FGF-2, hepatocyte growth factor, insulin-like growth factor 1 (IGF-I), and thymosin β4. Another group developed a prosurvival cocktail to boost the survival of myocardial grafts in the infarct rat heart (48). This cocktail was composed of Matrigel, a cell permeant peptide from B-cell lymphoma-extra large, cyclosporine A, a compound that opens ATP-dependent K channels, IGF-1 and ZVAD-fmk. More recently, IGF-1 and hepatocyte growth factor have shown to be able to improve LVEF and cell engraftment rate when delivered along with Sca-1+/CD31 cells in transplantation therapy (107). These studies highlight the modification of genes or extracellular microenvironment to modulate stem cell function and survival.

Conclusion and Future Perspectives

Stem cell therapy holds great promise to restore damaged heart and vessels. Researchers have made significant advancement in cell transplantation in preclinical and clinical settings. However, many problems still need to be resolved. Stem cell therapy for the treatment of ischemia tissues will require robust approaches for cell characterization, uniform isolation and/or maintenance, and optimal delivery strategies. Especially in cases where multiple cell types will be replaced and restored, long-term functional integration remains a challenge for effective clinical improvement. To address this limitation, bioengineering approaches to induce ordered multicellular architecture in vascular networks may be a key factor to enhance multicellular tissue integration and function (6). Furthermore, survival of transplanted cells or grafted tissue is still a challenge, considering the immunoreactive, ischemic, and necrotic host environment. Advancements in imaging capabilities will enable the ability to better track the location and survival of stem cells. Refining the delivery modality, dose, and phenotypic identify of the cells will also be critical to further enhance the functional effect of cell therapy. In the case of PSCs, concerns of teratoma formation will need to be mitigated by the use of highly purified, terminally differentiated, stem cell derivatives. In summary, the knowledge gained from past preclinical and clinical trials provides a wealth of information for further refinement of stem cell therapy for treating CVDs. Although future studies are needed to identify the optimal stem cell treatment for ischemic cardiovascular tissues, the hope of curative stem cell therapy remains bright.

GRANTS

This work was supported in part by National Institutes of Health Grants HL098688 and EB020235 (to N. F. Huang) and HL089315 and HL088957 (to YJW), Department of Veterans Affairs Biomedical Laboratory Research and Development Merit Review Award 1I01BX002310 (to NFH), the Stanford Chemistry Engineering & Medicine for Human Health (to NFH), the Stanford Cardiovascular Institute (to NFH), and a McCormick Gabilan fellowship (to NFH).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the author(s).

AUTHOR CONTRIBUTIONS

L.H. and N.F.H. conception and design of research; L.H., J.J.K., Y.J.W., and N.F.H. analyzed data; L.H., J.J.K., Y.J.W., and N.F.H. interpreted results of experiments; L.H. and N.F.H. prepared figures; L.H., J.J.K., and N.F.H. drafted manuscript; L.H., J.J.K., Y.J.W., and N.F.H. edited and revised manuscript; L.H., J.J.K., Y.J.W., and N.F.H. approved final version of manuscript.

REFERENCES

  • 1.Anokye-Danso F, Trivedi CM, Juhr D, Gupta M, Cui Z, Tian Y, Zhang Y, Yang W, Gruber PJ, Epstein JA, Morrisey EE. Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell 8: 376–388, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.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 275: 964–967, 1997. [DOI] [PubMed] [Google Scholar]
  • 3.Asahara T, Kawamoto A. Endothelial progenitor cells for postnatal vasculogenesis. Am J Physiol Cell Physiol 287: C572–C579, 2004. [DOI] [PubMed] [Google Scholar]
  • 4.Asahara T, Kawamoto A, Masuda H. Concise review: Circulating endothelial progenitor cells for vascular medicine. Stem Cells 29: 1650–1655, 2011. [DOI] [PubMed] [Google Scholar]
  • 5.Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert N, Grunwald F, Aicher A, Urbich C, Martin H, Hoelzer D, Dimmeler S, Zeiher AM. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (TOPCARE-AMI). Circulation 106: 3009–3017, 2002. [DOI] [PubMed] [Google Scholar]
  • 6.Baranski JD, Chaturvedi RR, Stevens KR, Eyckmans J, Carvalho B, Solorzano RD, Yang MT, Miller JS, Bhatia SN, Chen CS. Geometric control of vascular networks to enhance engineered tissue integration and function. Proc Natl Acad Sci USA 110: 7586–7591, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bernacki SH, Wall ME, Loboa EG. Isolation of human mesenchymal stem cells from bone and adipose tissue. Methods Cell Biol 86: 257–278, 2008. [DOI] [PubMed] [Google Scholar]
  • 8.Boilson BA, Kiernan TJ, Harbuzariu A, Nelson RE, Lerman A, Simari RD. Circulating CD34+ cell subsets in patients with coronary endothelial dysfunction. Nat Clin Pract Cardiovasc Med 5: 489–496, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Campagnoli C, Roberts IA, Kumar S, Bennett PR, Bellantuono I, Fisk NM. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood 98: 2396–2402, 2001. [DOI] [PubMed] [Google Scholar]
  • 10.Caplan AI. Mesenchymal stem cells. J Orthop Res 9: 641–650, 1991. [DOI] [PubMed] [Google Scholar]
  • 11.Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman ME, Capla JM, Galiano RD, Levine JP, Gurtner GC. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 10: 858–864, 2004. [DOI] [PubMed] [Google Scholar]
  • 12.Chen X, Aledia AS, Popson SA, Him L, Hughes CC, George SC. Rapid anastomosis of endothelial progenitor cell-derived vessels with host vasculature is promoted by a high density of cotransplanted fibroblasts. Tissue Eng Part A 16: 585–594, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cho SW, Moon SH, Lee SH, Kang SW, Kim J, Lim JM, Kim HS, Kim BS, Chung HM. Improvement of postnatal neovascularization by human embryonic stem cell derived endothelial-like cell transplantation in a mouse model of hindlimb ischemia. Circulation 116: 2409–2419, 2007. [DOI] [PubMed] [Google Scholar]
  • 14.Chong JJ, Yang X, Don CW, Minami E, Liu YW, Weyers JJ, Mahoney WM, Van Biber B, Cook SM, Palpant NJ, Gantz JA, Fugate JA, Muskheli V, Gough GM, Vogel KW, Astley CA, Hotchkiss CE, Baldessari A, Pabon L, Reinecke H, Gill EA, Nelson V, Kiem HP, Laflamme MA, Murry CE. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature 510: 273–277, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Cooke JP, Losordo DW. Modulating the vascular response to limb ischemia: angiogenic and cell therapies. Circ Res 116: 1561–1578, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Costa AR, Panda NC, Yong S, Mayorga ME, Pawlowski GP, Fan K, Penn MS, Laurita KR. Optical mapping of cryoinjured rat myocardium grafted with mesenchymal stem cells. Am J Physiol Heart Circ Physiol 302: H270–H277, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.De Haro J, Acin F, Lopez-Quintana A, Florez A, Martinez-Aguilar E, Varela C. Meta-analysis of randomized, controlled clinical trials in angiogenesis: gene and cell therapy in peripheral arterial disease. Heart Vessels 24: 321–328, 2009. [DOI] [PubMed] [Google Scholar]
  • 18.Deans RJ, Moseley AB. Mesenchymal stem cells: biology and potential clinical uses. Exp Hematol 28: 875–884, 2000. [DOI] [PubMed] [Google Scholar]
  • 19.Dormandy JA, Rutherford RB. Management of peripheral arterial disease (PAD). TASC Working Group TransAtlantic Inter-Society Consensus (TASC). J Vasc Surg 31: S1–S296, 2000. [PubMed] [Google Scholar]
  • 20.Gallo P, Peschle C, Condorelli G. Sources of cardiomyocytes for stem cell therapy: an update. Pediatr Res 59: 79R–83R, 2006. [DOI] [PubMed] [Google Scholar]
  • 21.Gersh BJ, Simari RD, Behfar A, Terzic CM, Terzic A. Cardiac cell repair therapy: a clinical perspective. Mayo Clin Proc 84: 876–892, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Gnecchi M, He H, Noiseux N, Liang OD, Zhang L, Morello F, Mu H, Melo LG, Pratt RE, Ingwall JS, Dzau VJ. Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J 20: 661–669, 2006. [DOI] [PubMed] [Google Scholar]
  • 23.Goumans MJ, Maring JA, Smits AM. A straightforward guide to the basic science behind cardiovascular cell-based therapies. Heart 100: 1153–1157, 2014. [DOI] [PubMed] [Google Scholar]
  • 24.Gu M, Nguyen PK, Lee AS, Xu D, Hu S, Plews JR, Han L, Huber BC, Lee WH, Gong Y, de Almeida PE, Lyons J, Ikeno F, Pacharinsak C, Connolly AJ, Gambhir SS, Robbins RC, Longaker MT, Wu JC. Microfluidic single-cell analysis shows that porcine induced pluripotent stem cell-derived endothelial cells improve myocardial function by paracrine activation. Circ Res 111: 882–893, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Han KH, Kim AK, Kim MH, Kim DH, Go HN, Kim DI. Enhancement of angiogenic effects by hypoxia-preconditioned human umbilical cord-derived mesenchymal stem cells in a mouse model of hindlimb ischemia. Cell Biol Int. 2015. July 29. doi: 10.1002/cbin.10519 [Epub ahead of print]. [DOI] [PubMed]
  • 26.Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP, Suncion VY, Tracy M, Ghersin E, Johnston PV, Brinker JA, Breton E, Davis-Sproul J, Schulman IH, Byrnes J, Mendizabal AM, Lowery MH, Rouy D, Altman P, Wong Po Foo C, Ruiz P, Amador A, Da Silva J, McNiece IK, Heldman AW, George R, Lardo A. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA 308: 2369–2379, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Heldman AW, DiFede DL, Fishman JE, Zambrano JP, Trachtenberg BH, Karantalis V, Mushtaq M, Williams AR, Suncion VY, McNiece IK, Ghersin E, Soto V, Lopera G, Miki R, Willens H, Hendel R, Mitrani R, Pattany P, Feigenbaum G, Oskouei B, Byrnes J, Lowery MH, Sierra J, Pujol MV, Delgado C, Gonzalez PJ, Rodriguez JE, Bagno LL, Rouy D, Altman P, Foo CW, da Silva J, Anderson E, Schwarz R, Mendizabal A, Hare JM. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC-HFT randomized trial. JAMA 311: 62–73, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Henderson JK, Draper JS, Baillie HS, Fishel S, Thomson JA, Moore H, Andrews PW. Preimplantation human embryos and embryonic stem cells show comparable expression of stage-specific embryonic antigens. Stem Cells 20: 329–337, 2002. [DOI] [PubMed] [Google Scholar]
  • 29.Hendrikx M, Hensen K, Clijsters C, Jongen H, Koninckx R, Bijnens E, Ingels M, Jacobs A, Geukens R, Dendale P, Vijgen J, Dilling D, Steels P, Mees U, Rummens JL. Recovery of regional but not global contractile function by the direct intramyocardial autologous bone marrow transplantation: results from a randomized controlled clinical trial. Circulation 114: I101–I107, 2006. [DOI] [PubMed] [Google Scholar]
  • 30.Hirsch AT, Criqui MH, Treat-Jacobson D, Regensteiner JG, Creager MA, Olin JW, Krook SH, Hunninghake DB, Comerota AJ, Walsh ME, McDermott MM, Hiatt WR. Peripheral arterial disease detection, awareness, and treatment in primary care. JAMA 286: 1317–1324, 2001. [DOI] [PubMed] [Google Scholar]
  • 31.Hirsch AT, Haskal ZJ, Hertzer NR, Bakal CW, Creager MA, Halperin JL, Hiratzka LF, Murphy WR, Olin JW, Puschett JB, Rosenfield KA, Sacks D, Stanley JC, Taylor LM Jr, White CJ, White J, White RA, Antman EM, Smith SC Jr, Adams CD, Anderson JL, Faxon DP, Fuster V, Gibbons RJ, Hunt SA, Jacobs AK, Nishimura R, Ornato JP, Page RL, Riegel B; American Association for Vascular Surgery; Society for Vascular Surgery; Society for Cardiovascular Angiography and Interventions; Society for Vascular Medicine and Biology; Society of Interventional Radiology; ACC/AHA Task Force on Practice Guidelines Writing Committee to Develop Guidelines for the Management of Patients With Peripheral Arterial Disease; American Association of Cardiovascular and Pulmonary Rehabilitation; National Heart, Lung, and Blood Institute; Society for Vascular Nursing; TransAtlantic Inter-Society Consensus; Vascular Disease Foundation . ACC/AHA 2005 Practice Guidelines for the management of patients with peripheral arterial disease (lower extremity, renal, mesenteric, and abdominal aortic): a collaborative report from the American Association for Vascular Surgery/Society for Vascular Surgery, Society for Cardiovascular Angiography and Interventions, Society for Vascular Medicine and Biology, Society of Interventional Radiology, and the ACC/AHA Task Force on Practice Guidelines (Writing Committee to Develop Guidelines for the Management of Patients With Peripheral Arterial Disease): endorsed by the American Association of Cardiovascular and Pulmonary Rehabilitation; National Heart, Lung, and Blood Institute; Society for Vascular Nursing; TransAtlantic Inter-Society Consensus; and Vascular Disease Foundation. Circulation 113: e463–e654, 2006. [DOI] [PubMed] [Google Scholar]
  • 32.Hodgson DM, Behfar A, Zingman LV, Kane GC, Perez-Terzic C, Alekseev AE, Puceat M, Terzic A. Stable benefit of embryonic stem cell therapy in myocardial infarction. Am J Physiol Heart Circ Physiol 287: H471–H479, 2004. [DOI] [PubMed] [Google Scholar]
  • 33.Hoover-Plow J, Gong Y. Challenges for heart disease stem cell therapy. Vasc Health Risk Manag 8: 99–113, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hristov M, Erl W, Weber PC. Endothelial progenitor cells: mobilization, differentiation, and homing. Arterioscler Thromb Vasc Biol 23: 1185–1189, 2003. [DOI] [PubMed] [Google Scholar]
  • 35.Huang NF, Niiyama H, De A, Gambhir SS, Cooke JP. Embryonic stem cell-derived endothelial cells for treatment of hindlimb ischemia. J Vis Exp 23: pii: 1034, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Huang NF, Chu J, Lee RJ, Li S. Biophysical and chemical effects of fibrin on mesenchymal stromal cell gene expression. Acta Biomater 6: 3947–3956, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Huang NF, Niiyama H, Peter C, De A, Natkunam Y, Fleissner F, Li Z, Rollins MD, Wu JC, Gambhir SS, Cooke JP. Embryonic stem cell-derived endothelial cells engraft into the ischemic hindlimb and restore perfusion. Arterioscler Thromb Vasc Biol 30: 984–991, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Huang P, Li S, Han M, Xiao Z, Yang R, Han ZC. Autologous transplantation of granulocyte colony-stimulating factor-mobilized peripheral blood mononuclear cells improves critical limb ischemia in diabetes. Diabetes Care 28: 2155–2160, 2005. [DOI] [PubMed] [Google Scholar]
  • 39.Iafrati MD, Hallett JW, Geils G, Pearl G, Lumsden A, Peden E, Bandyk D, Vijayaraghava KS, Radhakrishnan R, Ascher E, Hingorani A, Roddy S. Early results and lessons learned from a multicenter, randomized, double-blind trial of bone marrow aspirate concentrate in critical limb ischemia. J Vasc Surg 54: 1650–1658, 2011. [DOI] [PubMed] [Google Scholar]
  • 40.Iwase T, Nagaya N, Fujii T, Itoh T, Murakami S, Matsumoto T, Kangawa K, Kitamura S. Comparison of angiogenic potency between mesenchymal stem cells and mononuclear cells in a rat model of hindlimb ischemia. Cardiovasc Res 66: 543–551, 2005. [DOI] [PubMed] [Google Scholar]
  • 41.Jeon O, Song SJ, Bhang SH, Choi CY, Kim MJ, Kim BS. Additive effect of endothelial progenitor cell mobilization and bone marrow mononuclear cell transplantation on angiogenesis in mouse ischemic limbs. J Biomed Sci 14: 323–330, 2007. [DOI] [PubMed] [Google Scholar]
  • 42.Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, Du J, Aldrich S, Lisberg A, Low WC, Largaespada DA, Verfaillie CM. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418: 41–49, 2002. [DOI] [PubMed] [Google Scholar]
  • 43.Kamihata H, Matsubara H, Nishiue T, Fujiyama S, Amano K, Iba O, Imada T, Iwasaka T. Improvement of collateral perfusion and regional function by implantation of peripheral blood mononuclear cells into ischemic hibernating myocardium. Arterioscler Thromb Vasc Biol 22: 1804–1810, 2002. [DOI] [PubMed] [Google Scholar]
  • 44.Kikuchi K, Poss KD. Cardiac regenerative capacity and mechanisms. Annu Rev Cell Dev Biol 28: 719–741, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kinnaird T, Stabile E, Burnett MS, Lee CW, Barr S, Fuchs S, Epstein SE. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circ Res 94: 678–685, 2004. [DOI] [PubMed] [Google Scholar]
  • 46.Kinnaird T, Stabile E, Burnett MS, Shou M, Lee CW, Barr S, Fuchs S, Epstein SE. Local delivery of marrow-derived stromal cells augments collateral perfusion through paracrine mechanisms. Circulation 109: 1543–1549, 2004. [DOI] [PubMed] [Google Scholar]
  • 47.Kobayashi K, Kondo T, Inoue N, Aoki M, Mizuno M, Komori K, Yoshida J, Murohara T. Combination of in vivo angiopoietin-1 gene transfer and autologous bone marrow cell implantation for functional therapeutic angiogenesis. Arterioscler Thromb Vasc Biol 26: 1465–1472, 2006. [DOI] [PubMed] [Google Scholar]
  • 48.Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA, Dupras SK, Reinecke H, Xu C, Hassanipour M, Police S, O'Sullivan C, Collins L, Chen Y, Minami E, Gill EA, Ueno S, Yuan C, Gold J, Murry CE. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat Biotechnol 25: 1015–1024, 2007. [DOI] [PubMed] [Google Scholar]
  • 49.Li H, Zuo S, He Z, Yang Y, Pasha Z, Wang Y, Xu M. Paracrine factors released by GATA-4 overexpressed mesenchymal stem cells increase angiogenesis and cell survival. Am J Physiol Heart Circ Physiol 299: H1772–H1781, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Li Z, Wu JC, Sheikh AY, Kraft D, Cao F, Xie X, Patel M, Gambhir SS, Robbins RC, Cooke JP, Wu JC. Differentiation, survival, and function of embryonic stem cell derived endothelial cells for ischemic heart disease. Circulation 116: I46–I54, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lian X, Bao X, Al-Ahmad A, Liu J, Wu Y, Dong W, Dunn KK, Shusta EV, Palecek SP. Efficient differentiation of human pluripotent stem cells to endothelial progenitors via small-molecule activation of WNT signaling. Stem Cell Reports 3: 804–816, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Lister R, Pelizzola M, Kida YS, Hawkins RD, Nery JR, Hon G, Antosiewicz-Bourget J, O'Malley R, Castanon R, Klugman S, Downes M, Yu R, Stewart R, Ren B, Thomson JA, Evans RM, Ecker JR. Hotspots of aberrant epigenomic reprogramming in human induced pluripotent stem cells. Nature 471: 68–73, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Losordo DW, Henry TD, Davidson C, Sup Lee J, Costa MA, Bass T, Mendelsohn F, Fortuin FD, Pepine CJ, Traverse JH, Amrani D, Ewenstein BM, Riedel N, Story K, Barker K, Povsic TJ, Harrington RA, Schatz RA, Investigators AC . Intramyocardial, autologous CD34+ cell therapy for refractory angina. Circ Res 109: 428–436, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lu D, Chen B, Liang Z, Deng W, Jiang Y, Li S, Xu J, Wu Q, Zhang Z, Xie B, Chen S. Comparison of bone marrow mesenchymal stem cells with bone marrow-derived mononuclear cells for treatment of diabetic critical limb ischemia and foot ulcer: a double-blind, randomized, controlled trial. Diabetes Res Clin Pract 92: 26–36, 2011. [DOI] [PubMed] [Google Scholar]
  • 55.Makkar RR, Smith RR, Cheng K, Malliaras K, Thomson LE, Berman D, Czer LS, Marban L, Mendizabal A, Johnston PV, Russell SD, Schuleri KH, Lardo AC, Gerstenblith G, Marban E. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet 379: 895–904, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.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 9: 1195–1201, 2003. [DOI] [PubMed] [Google Scholar]
  • 57.Menard C, Hagege AA, Agbulut O, Barro M, Morichetti MC, Brasselet C, Bel A, Messas E, Bissery A, Bruneval P, Desnos M, Puceat M, Menasche P. Transplantation of cardiac-committed mouse embryonic stem cells to infarcted sheep myocardium: a preclinical study. Lancet 366: 1005–1012, 2005. [DOI] [PubMed] [Google Scholar]
  • 58.Menasche P, Hagege AA, Vilquin JT, Desnos M, Abergel E, Pouzet B, Bel A, Sarateanu S, Scorsin M, Schwartz K, Bruneval P, Benbunan M, Marolleau JP, Duboc D. Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction. J Am Coll Cardiol 41: 1078–1083, 2003. [DOI] [PubMed] [Google Scholar]
  • 59.Menasche P, Alfieri O, Janssens S, McKenna W, Reichenspurner H, Trinquart L, Vilquin JT, Marolleau JP, Seymour B, Larghero J, Lake S, Chatellier G, Solomon S, Desnos M, Hagege AA. The Myoblast Autologous Grafting in Ischemic Cardiomyopathy (MAGIC) trial: first randomized placebo-controlled study of myoblast transplantation. Circulation 117: 1189–1200, 2008. [DOI] [PubMed] [Google Scholar]
  • 60.Michael LH, Entman ML, Hartley CJ, Youker KA, Zhu J, Hall SR, Hawkins HK, Berens K, Ballantyne CM. Myocardial ischemia and reperfusion: a murine model. Am J Physiol Heart Circ Physiol 269: H2147–H2154, 1995. [DOI] [PubMed] [Google Scholar]
  • 61.Miyahara Y, Nagaya N, Kataoka M, Yanagawa B, Tanaka K, Hao H, Ishino K, Ishida H, Shimizu T, Kangawa K, Sano S, Okano T, Kitamura S, Mori H. Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction. Nat Med 12: 459–465, 2006. [DOI] [PubMed] [Google Scholar]
  • 62.Mohammadzadeh L, Samedanifard SH, Keshavarzi A, Alimoghaddam K, Larijani B, Ghavamzadeh A, Ahmadi AS, Shojaeifard A, Ostadali MR, Sharifi AM, Amini MR, Mahmoudian A, Fakhraei H, Aalaa M, Mohajeri-Tehrani MR. Therapeutic outcomes of transplanting autologous granulocyte colony-stimulating factor-mobilised peripheral mononuclear cells in diabetic patients with critical limb ischaemia. Exp Clin Endocrinol Diabetes 121: 48–53, 2013. [DOI] [PubMed] [Google Scholar]
  • 63.Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S, Despres J, Fullerton HJ, Howard VJ, Huffman MD, Judd SE, Kissela BM, Lackland DT, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Matchar DB, McGuire DK, Mohler ER, Moy CS 3rd, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Willey JZ, Woo D, Yeh RW, Turner MB. Heart disease and stroke statistics–2015 update: a report from the American Heart Association. Circulation 131: e29–e322, 2015. [DOI] [PubMed] [Google Scholar]
  • 64.Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S, Després JP, Fullerton HJ, Howard VJ, Huffman MD, Judd SE, Kissela BM, Lackland DT, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Matchar DB, McGuire DK, Mohler ER 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Willey JZ, Woo D, Yeh RW, Turner MB; American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics–2015 update: a report from the American Heart Association. Circulation 131: e29–e322, 2015. [DOI] [PubMed] [Google Scholar]
  • 65.Muller-Ehmsen J, Whittaker P, Kloner RA, Dow JS, Sakoda T, Long TI, Laird PW, Kedes L. Survival and development of neonatal rat cardiomyocytes transplanted into adult myocardium. J Mol Cell Cardiol 34: 107–116, 2002. [DOI] [PubMed] [Google Scholar]
  • 66.Mureli S, Gans CP, Bare DJ, Geenen DL, Kumar NM, Banach K. Mesenchymal stem cells improve cardiac conduction by upregulation of connexin 43 through paracrine signaling. Am J Physiol Heart Circ Physiol 304: H600–H609, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Niiyama H, Huang NF, Rollins MD, Cooke JP. Murine model of hindlimb ischemia. J Vis Exp 23: pii: 1035, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R, Nadal-Ginard B, Bodine DM, Leri A, Anversa P. Bone marrow cells regenerate infarcted myocardium. Nature 410: 701–705, 2001. [DOI] [PubMed] [Google Scholar]
  • 69.Oswald J, Boxberger S, Jorgensen B, Feldmann S, Ehninger G, Bornhauser M, Werner C. Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Stem Cells 22: 377–384, 2004. [DOI] [PubMed] [Google Scholar]
  • 70.Ouriel K. Peripheral arterial disease. Lancet 358: 1257–1264, 2001. [DOI] [PubMed] [Google Scholar]
  • 71.Ozturk A, Kucukardali Y, Tangi F, Erikci A, Uzun G, Bashekim C, Sen H, Terekeci H, Narin Y, Ozyurt M, Ozkan S, Sayan O, Rodop O, Nalbant S, Sildiroglu O, Yalniz FF, Senkal IV, Sabuncu H, Oktenli C. Therapeutical potential of autologous peripheral blood mononuclear cell transplantation in patients with type 2 diabetic critical limb ischemia. J Diabetes Complications 26: 29–33, 2012. [DOI] [PubMed] [Google Scholar]
  • 72.Perin EC, Willerson JT, Pepine CJ, Henry TD, Ellis SG, Zhao DX, Silva GV, Lai D, Thomas JD, Kronenberg MW, Martin AD, Anderson RD, Traverse JH, Penn MS, Anwaruddin S, Hatzopoulos AK, Gee AP, Taylor DA, Cogle CR, Smith D, Westbrook L, Chen J, Handberg E, Olson RE, Geither C, Bowman S, Francescon J, Baraniuk S, Piller LB, Simpson LM, Loghin C, Aguilar D, Richman S, Zierold C, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moyé LA, Simari RD; Cardiovascular Cell Therapy Research Network (CCTRN) . Effect of transendocardial delivery of autologous bone marrow mononuclear cells on functional capacity, left ventricular function, and perfusion in chronic heart failure: the FOCUS-CCTRN trial. JAMA 307: 1717–1726, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science 284: 143–147, 1999. [DOI] [PubMed] [Google Scholar]
  • 74.Pittenger MF, Martin BJ. Mesenchymal stem cells and their potential as cardiac therapeutics. Circ Res 95: 9–20, 2004. [DOI] [PubMed] [Google Scholar]
  • 75.Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 276: 71–74, 1997. [DOI] [PubMed] [Google Scholar]
  • 76.Raval Z, Losordo DW. Cell therapy of peripheral arterial disease: from experimental findings to clinical trials. Circ Res 112: 1288–1302, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Reinecke H, Zhang M, Bartosek T, Murry CE. Survival, integration, and differentiation of cardiomyocyte grafts: a study in normal and injured rat hearts. Circulation 100: 193–202, 1999. [DOI] [PubMed] [Google Scholar]
  • 78.Resch T, Pircher A, Kahler CM, Pratschke J, Hilbe W. Endothelial progenitor cells: current issues on characterization and challenging clinical applications. Stem Cell Rev 8: 926–939, 2012. [DOI] [PubMed] [Google Scholar]
  • 79.Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A. Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol 18: 399–404, 2000. [DOI] [PubMed] [Google Scholar]
  • 80.Rhee YH, Ko JY, Chang MY, Yi SH, Kim D, Kim CH, Shim JW, Jo AY, Kim BW, Lee H, Lee SH, Suh W, Park CH, Koh HC, Lee YS, Lanza R, Kim KS, Lee SH. Protein-based human iPS cells efficiently generate functional dopamine neurons and can treat a rat model of Parkinson disease. J Clin Invest 121: 2326–2335, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Roger VL, Go AS, Lloyd-Jones DM, Benjamin EJ, Berry JD, Borden WB, Bravata DM, Dai S, Ford ES, Fox CS, Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, Howard VJ, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Makuc DM, Marcus GM, Marelli A, Matchar DB, Moy CS, Mozaffarian D, Mussolino ME, Nichol G, Paynter NP, Soliman EZ, Sorlie PD, Sotoodehnia N, Turan TN, Virani SS, Wong ND, Woo D, Turner MB; American Heart Association Statistics Committee Stroke Statistics Subcommittee. Executive summary: heart disease and stroke statistics–2012 update: a report from the American Heart Association. Circulation 125: 188–197, 2012. [DOI] [PubMed] [Google Scholar]
  • 82.Ruan W, Pan CZ, Huang GQ, Li YL, Ge JB, Shu XH. Assessment of left ventricular segmental function after autologous bone marrow stem cells transplantation in patients with acute myocardial infarction by tissue tracking and strain imaging. Chin Med J (Engl) 118: 1175–1181, 2005. [PubMed] [Google Scholar]
  • 83.Rufaihah AJ, Huang NF, Jame S, Lee JC, Nguyen HN, Byers B, De A, Okogbaa J, Rollins M, Reijo-Pera R, Gambhir SS, Cooke JP. Endothelial cells derived from human iPSCS increase capillary density and improve perfusion in a mouse model of peripheral arterial disease. Arterioscler Thromb Vasc Biol 31: e72–e79, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Schachinger V, Erbs S, Elsasser A, Haberbosch W, Hambrecht R, Holschermann H, Yu J, Corti R, Mathey DG, Hamm CW, Suselbeck T, Assmus B, Tonn T, Dimmeler S, Zeiher AM, Investigators RA . Intracoronary bone marrow-derived progenitor cells in acute myocardial infarction. N Engl J Med 355: 1210–1221, 2006. [DOI] [PubMed] [Google Scholar]
  • 85.Seeger FH, Tonn T, Krzossok N, Zeiher AM, Dimmeler S. Cell isolation procedures matter: a comparison of different isolation protocols of bone marrow mononuclear cells used for cell therapy in patients with acute myocardial infarction. Eur Heart J 28: 766–772, 2007. [DOI] [PubMed] [Google Scholar]
  • 86.Segers VF, Lee RT. Stem-cell therapy for cardiac disease. Nature 451: 937–942, 2008. [DOI] [PubMed] [Google Scholar]
  • 87.Shake JG, Gruber PJ, Baumgartner WA, Senechal G, Meyers J, Redmond JM, Pittenger MF, Martin BJ. Mesenchymal stem cell implantation in a swine myocardial infarct model: engraftment and functional effects. Ann Thorac Surg 73: 1919–1925, 2002. [DOI] [PubMed] [Google Scholar]
  • 88.Shintani S, Murohara T, Ikeda H, Ueno T, Sasaki K, Duan J, Imaizumi T. Augmentation of postnatal neovascularization with autologous bone marrow transplantation. Circulation 103: 897–903, 2001. [DOI] [PubMed] [Google Scholar]
  • 89.Simmons PJ, Torok-Storb B. Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood 78: 55–62, 1991. [PubMed] [Google Scholar]
  • 90.Suuronen EJ, Veinot JP, Wong S, Kapila V, Price J, Griffith M, Mesana TG, Ruel M. Tissue-engineered injectable collagen-based matrices for improved cell delivery and vascularization of ischemic tissue using CD133+ progenitors expanded from the peripheral blood. Circulation 114: I138–I144, 2006. [DOI] [PubMed] [Google Scholar]
  • 91.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126: 663–676, 2006. [DOI] [PubMed] [Google Scholar]
  • 92.Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131: 861–872, 2007. [DOI] [PubMed] [Google Scholar]
  • 93.Takeshita S, Zheng LP, Brogi E, Kearney M, Pu LQ, Bunting S, Ferrara N, Symes JF, Isner JM. Therapeutic angiogenesis. A single intraarterial bolus of vascular endothelial growth factor augments revascularization in a rabbit ischemic hind limb model. J Clin Invest 93: 662–670, 1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S, Masaki H, Amano K, Kishimoto Y, Yoshimoto K, Akashi H, Shimada K, Iwasaka T, Imaizumi T; Therapeutic Angiogenesis using Cell Transplantation (TACT) Study Investigators. Therapeutic angiogenesis for patients with limb ischaemia by autologous transplantation of bone-marrow cells: a pilot study and a randomised controlled trial. Lancet 360: 427–435, 2002. [DOI] [PubMed] [Google Scholar]
  • 95.Telukuntla KS, Suncion VY, Schulman IH, Hare JM. The advancing field of cell-based therapy: insights and lessons from clinical trials. J Am Heart Assoc 2: e000338, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Templin C, Zweigerdt R, Schwanke K, Olmer R, Ghadri JR, Emmert MY, Muller E, Kuest SM, Cohrs S, Schibli R, Kronen P, Hilbe M, Reinisch A, Strunk D, Haverich A, Hoerstrup S, Luscher TF, Kaufmann PA, Landmesser U, Martin U. Transplantation and tracking of human-induced pluripotent stem cells in a pig model of myocardial infarction: assessment of cell survival, engraftment, and distribution by hybrid single photon emission computed tomography/computed tomography of sodium iodide symporter transgene expression. Circulation 126: 430–439, 2012. [DOI] [PubMed] [Google Scholar]
  • 97.Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science 282: 1145–1147, 1998. [DOI] [PubMed] [Google Scholar]
  • 98.Thum T, Tsikas D, Stein S, Schultheiss M, Eigenthaler M, Anker SD, Poole-Wilson PA, Ertl G, Bauersachs J. Suppression of endothelial progenitor cells in human coronary artery disease by the endogenous nitric oxide synthase inhibitor asymmetric dimethylarginine. J Am Coll Cardiol 46: 1693–1701, 2005. [DOI] [PubMed] [Google Scholar]
  • 99.Traverse JH, Henry TD, Ellis SG, Pepine CJ, Willerson JT, Zhao DX, Forder JR, Byrne BJ, Hatzopoulos AK, Penn MS, Perin EC, Baran KW, Chambers J, Lambert C, Raveendran G, Simon DI, Vaughan DE, Simpson LM, Gee AP, Taylor DA, Cogle CR, Thomas JD, Silva GV, Jorgenson BC, Olson RE, Bowman S, Francescon J, Geither C, Handberg E, Smith DX, Baraniuk S, Piller LB, Loghin C, Aguilar D, Richman S, Zierold C, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moyé LA, Simari RD; Cardiovascular Cell Therapy ResearchNetwork. Effect of intracoronary delivery of autologous bone marrow mononuclear cells 2 to 3 weeks following acute myocardial infarction on left ventricular function: the Late TIME randomized trial. JAMA 306: 2110–2119, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Traverse JH, Henry TD, Pepine CJ, Willerson JT, Zhao DX, Ellis SG, Forder JR, Anderson RD, Hatzopoulos AK, Penn MS, Perin EC, Chambers J, Baran KW, Raveendran G, Lambert C, Lerman A, Simon DI, Vaughan DE, Lai D, Gee AP, Taylor DA, Cogle CR, Thomas JD, Olson RE, Bowman S, Francescon J, Geither C, Handberg E, Kappenman C, Westbrook L, Piller LB, Simpson LM, Baraniuk S, Loghin C, Aguilar D, Richman S, Zierold C, Spoon DB, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moyé LA, Simari RD; Cardiovascular Cell Therapy Research Network (CCTRN) . Effect of the use and timing of bone marrow mononuclear cell delivery on left ventricular function after acute myocardial infarction: the TIME randomized trial. JAMA 308: 2380–2389, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Van Laake LW, Van Hoof D, Mummery CL. Cardiomyocytes derived from stem cells. Ann Med 37: 499–512, 2005. [DOI] [PubMed] [Google Scholar]
  • 102.van Laake LW, Hassink R, Doevendans PA, Mummery C. Heart repair and stem cells. J Physiol 577: 467–478, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Van Tongeren RB, Hamming JF, Fibbe WE, Van Weel V, Frerichs SJ, Stiggelbout AM, Van Bockel JH, Lindeman JH. Intramuscular or combined intramuscular/intra-arterial administration of bone marrow mononuclear cells: a clinical trial in patients with advanced limb ischemia. J Cardiovasc Surg (Torino) 49: 51–58, 2008. [PubMed] [Google Scholar]
  • 104.Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM, Dimmeler S. Number and migratory activity of circulating endothelial progenitor cells inversely correlate with risk factors for coronary artery disease. Circ Res 89: E1–E7, 2001. [DOI] [PubMed] [Google Scholar]
  • 105.Walter DH, Krankenberg H, Balzer JO, Kalka C, Baumgartner I, Schluter M, Tonn T, Seeger F, Dimmeler S, Lindhoff-Last E, Zeiher AM, Investigators P . Intraarterial administration of bone marrow mononuclear cells in patients with critical limb ischemia: a randomized-start, placebo-controlled pilot trial (PROVASA). Circ Cardiovasc Interv 4: 26–37, 2011. [DOI] [PubMed] [Google Scholar]
  • 106.Wang H, Riha GM, Yan S, Li M, Chai H, Yang H, Yao Q, Chen C. Shear stress induces endothelial differentiation from a murine embryonic mesenchymal progenitor cell line. Arterioscler Thromb Vasc Biol 25: 1817–1823, 2005. [DOI] [PubMed] [Google Scholar]
  • 107.Wang X, Li Q, Hu Q, Suntharalingam P, From AH, Zhang J. Intra-myocardial injection of both growth factors and heart derived Sca-1+/CD31- cells attenuates post-MI LV remodeling more than does cell transplantation alone: neither intervention enhances functionally significant cardiomyocyte regeneration. PLoS One 9: e95247, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.White MP, Rufaihah AJ, Liu L, Ghebremariam YT, Ivey KN, Cooke JP, Srivastava D. Limited gene expression variation in human embryonic stem cell and induced pluripotent stem cell-derived endothelial cells. Stem Cells 31: 92–103, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Wingate K, Bonani W, Tan Y, Bryant SJ, Tan W. Compressive elasticity of three-dimensional nanofiber matrix directs mesenchymal stem cell differentiation to vascular cells with endothelial or smooth muscle cell markers. Acta Biomater 8: 1440–1449, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Wingate K, Floren M, Tan Y, Tseng PO, Tan W. Synergism of matrix stiffness and vascular endothelial growth factor on mesenchymal stem cells for vascular endothelial regeneration. Tissue Eng Part A 20: 2503–2512, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Wobus AM, Boheler KR. Embryonic stem cells: prospects for developmental biology and cell therapy. Physiol Rev 85: 635–678, 2005. [DOI] [PubMed] [Google Scholar]
  • 112.Wollert KC, Meyer GP, Lotz J, Ringes-Lichtenberg S, Lippolt P, Breidenbach C, Fichtner S, Korte T, Hornig B, Messinger D, Arseniev L, Hertenstein B, Ganser A, Drexler H. Intracoronary autologous bone-marrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial. Lancet 364: 141–148, 2004. [DOI] [PubMed] [Google Scholar]
  • 113.Xiong Q, Hill KL, Li Q, Suntharalingam P, Mansoor A, Wang X, Jameel MN, Zhang P, Swingen C, Kaufman DS, Zhang J. A fibrin patch-based enhanced delivery of human embryonic stem cell-derived vascular cell transplantation in a porcine model of postinfarction left ventricular remodeling. Stem Cells 29: 367–375, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Xu C, Inokuma MS, Denham J, Golds K, Kundu P, Gold JD, Carpenter MK. Feeder-free growth of undifferentiated human embryonic stem cells. Nat Biotechnol 19: 971–974, 2001. [DOI] [PubMed] [Google Scholar]
  • 115.Xu Y, Meng H, Li C, Hao M, Wang Y, Yu Z, Li Q, Han J, Zhai Q, Qiu L. Umbilical cord-derived mesenchymal stem cells isolated by a novel explantation technique can differentiate into functional endothelial cells and promote revascularization. Stem Cells Dev 19: 1511–1522, 2010. [DOI] [PubMed] [Google Scholar]
  • 116.Yamada S, Nelson TJ, Crespo-Diaz RJ, Perez-Terzic C, Liu XK, Miki T, Seino S, Behfar A, Terzic A. Embryonic stem cell therapy of heart failure in genetic cardiomyopathy. Stem Cells 26: 2644–2653, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Yamahara K, Sone M, Itoh H, Yamashita JK, Yurugi-Kobayashi T, Homma K, Chao TH, Miyashita K, Park K, Oyamada N, Sawada N, Taura D, Fukunaga Y, Tamura N, Nakao K. Augmentation of neovascularization [corrected] in hindlimb ischemia by combined transplantation of human embryonic stem cells-derived endothelial and mural cells. PLoS One 3: e1666, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Yamashita J, Itoh H, Hirashima M, Ogawa M, Nishikawa S, Yurugi T, Naito M, Nakao K, Nishikawa S. Flk1-positive cells derived from embryonic stem cells serve as vascular progenitors. Nature 408: 92–96, 2000. [DOI] [PubMed] [Google Scholar]
  • 119.Zhang H, Zhang N, Li M, Feng H, Jin W, Zhao H, Chen X, Tian L. Therapeutic angiogenesis of bone marrow mononuclear cells (MNCs) and peripheral blood MNCs: transplantation for ischemic hindlimb. Ann Vasc Surg 22: 238–247, 2008. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Heart and Circulatory Physiology are provided here courtesy of American Physiological Society

RESOURCES