Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 120(9): 1466-1476, 2017 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-28298297

RESUMEN

RATIONALE: Paracrine secretions seem to mediate therapeutic effects of human CD34+ stem cells locally transplanted in patients with myocardial and critical limb ischemia and in animal models. Earlier, we had discovered that paracrine secretion from human CD34+ cells contains proangiogenic, membrane-bound nanovesicles called exosomes (CD34Exo). OBJECTIVE: Here, we investigated the mechanisms of CD34Exo-mediated ischemic tissue repair and therapeutic angiogenesis by studying their miRNA content and uptake. METHODS AND RESULTS: When injected into mouse ischemic hindlimb tissue, CD34Exo, but not the CD34Exo-depleted conditioned media, mimicked the beneficial activity of their parent cells by improving ischemic limb perfusion, capillary density, motor function, and their amputation. CD34Exo were found to be enriched with proangiogenic miRNAs such as miR-126-3p. Knocking down miR-126-3p from CD34Exo abolished their angiogenic activity and beneficial function both in vitro and in vivo. Interestingly, injection of CD34Exo increased miR-126-3p levels in mouse ischemic limb but did not affect the endogenous synthesis of miR-126-3p, suggesting a direct transfer of stable and functional exosomal miR-126-3p. miR-126-3p enhanced angiogenesis by suppressing the expression of its known target, SPRED1, simultaneously modulating the expression of genes involved in angiogenic pathways such as VEGF (vascular endothelial growth factor), ANG1 (angiopoietin 1), ANG2 (angiopoietin 2), MMP9 (matrix metallopeptidase 9), TSP1 (thrombospondin 1), etc. Interestingly, CD34Exo, when treated to ischemic hindlimbs, were most efficiently internalized by endothelial cells relative to smooth muscle cells and fibroblasts, demonstrating a direct role of stem cell-derived exosomes on mouse endothelium at the cellular level. CONCLUSIONS: Collectively, our results have demonstrated a novel mechanism by which cell-free CD34Exo mediates ischemic tissue repair via beneficial angiogenesis. Exosome-shuttled proangiogenic miRNAs may signify amplification of stem cell function and may explain the angiogenic and therapeutic benefits associated with CD34+ stem cell therapy.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Antígenos CD34/metabolismo , Células Progenitoras Endoteliales/trasplante , Exosomas/trasplante , Isquemia/cirugía , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Proteínas Angiogénicas/genética , Animales , Biomarcadores/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/metabolismo , Exosomas/metabolismo , Regulación de la Expresión Génica , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Isquemia/genética , Isquemia/metabolismo , Isquemia/fisiopatología , Ratones Endogámicos BALB C , MicroARNs/genética , MicroARNs/metabolismo , Actividad Motora , Comunicación Paracrina , Fenotipo , Interferencia de ARN , Recuperación de la Función , Flujo Sanguíneo Regional , Transducción de Señal , Factores de Tiempo , Transfección
2.
J Mol Cell Cardiol ; 74: 231-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25009075

RESUMEN

The translation of cell-based therapies for ischemic tissue repair remains limited by several factors, including poor cell survival and limited target site retention. Advances in nanotechnology enable the development of specifically designed delivery matrices to address these limitations and thereby improve the efficacy of cell-based therapies. Given the relevance of integrin signaling for cellular homeostasis, we developed an injectable, bioactive peptide-based nanofiber matrix that presents an integrin-binding epitope derived from fibronectin, and evaluated its feasibility as a supportive artificial matrix for bone marrow-derived pro-angiogenic cells (BMPACs) used as a therapy in ischemic tissue repair. Incubation of BMPACs with these peptide nanofibers in vitro significantly attenuated apoptosis while enhancing proliferation and adhesion. Pro-angiogenic function was enhanced, as cells readily formed tubes. These effects were, in part, mediated via p38, and p44/p42 MAP kinases, which are downstream pathways of focal adhesion kinase. In a murine model of hind limb ischemia, an intramuscular injection of BMPACs within this bioactive peptide nanofiber matrix resulted in greater retention of cells, enhanced capillary density, increased limb perfusion, reduced necrosis/amputation, and preserved function of the ischemic limb compared to treatment with cells alone. This self-assembling, bioactive peptide nanofiber matrix presenting an integrin-binding domain of fibronectin improves regenerative efficacy of cell-based strategies in ischemic tissue by enhancing cell survival, retention, and reparative functions.


Asunto(s)
Células de la Médula Ósea/citología , Epítopos/metabolismo , Fibronectinas/metabolismo , Isquemia/terapia , Nanofibras/administración & dosificación , Péptidos/administración & dosificación , Animales , Materiales Biocompatibles , Células de la Médula Ósea/metabolismo , Supervivencia Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Epítopos/química , Fibronectinas/química , Expresión Génica , Miembro Posterior/irrigación sanguínea , Miembro Posterior/efectos de los fármacos , Miembro Posterior/lesiones , Integrinas/metabolismo , Isquemia/patología , Masculino , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Nanofibras/química , Neovascularización Fisiológica , Péptidos/síntesis química , Péptidos/metabolismo , Unión Proteica , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
J Biol Chem ; 288(25): 18022-34, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23645678

RESUMEN

We have shown previously that estrogen (estradiol, E2) supplementation enhances voluntary alcohol consumption in ovariectomized female rodents and that increased alcohol consumption impairs ischemic hind limb vascular repair. However, the effect of E2-induced alcohol consumption on post-infarct myocardial repair and on the phenotypic/functional properties of endothelial progenitor cells (EPCs) is not known. Additionally, the molecular signaling of alcohol-estrogen interactions remains to be elucidated. This study examined the effect of E2-induced increases in ethanol consumption on post-infarct myocardial function/repair. Ovariectomized female mice, implanted with 17ß-E2 or placebo pellets were given access to alcohol for 6 weeks and subjected to acute myocardial infarction. Left ventricular functions were consistently depressed in mice consuming ethanol compared with those receiving only E2. Alcohol-consuming mice also displayed significantly increased infarct size and reduced capillary density. Ethanol consumption also reduced E2-induced mobilization and homing of EPCs to injured myocardium compared with the E2-alone group. In vitro, exposure of EPCs to ethanol suppressed E2-induced proliferation, survival, and migration and markedly altered E2-induced estrogen receptor-dependent cell survival signaling and gene expression. Furthermore, ethanol-mediated suppression of EPC biology was endothelial nitric oxide synthase-dependent because endothelial nitric oxide synthase-null mice displayed an exaggerated response to post-acute myocardial infarction left ventricular functions. These data suggest that E2 modulation of alcohol consumption, and the ensuing EPC dysfunction, may negatively compete with the beneficial effects of estrogen on post-infarct myocardial repair.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Estradiol/farmacología , Etanol/farmacología , Miocardio/metabolismo , Células Madre/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Estradiol/metabolismo , Estrógenos/metabolismo , Estrógenos/farmacología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/patología , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Ovariectomía , Unión Proteica/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Células Madre/metabolismo , Células Madre/fisiología
4.
Circulation ; 127(1): 63-73, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23204107

RESUMEN

BACKGROUND: CXC-chemokine receptor 4 (CXCR4) regulates the retention of stem/progenitor cells in the bone marrow (BM), and the CXCR4 antagonist AMD3100 improves recovery from coronary ligation injury by mobilizing stem/progenitor cells from the BM to the peripheral blood. Thus, we investigated whether AMD3100 also improves recovery from ischemia/reperfusion injury, which more closely mimics myocardial infarction in patients, because blood flow is only temporarily obstructed. METHODS AND RESULTS: Mice were treated with single subcutaneous injections of AMD3100 (5 mg/kg) or saline after ischemia/reperfusion injury. Three days later, histological measurements of the ratio of infarct area to area at risk were smaller in AMD3100-treated mice than in mice administered saline, and echocardiographic measurements of left ventricular function were greater in the AMD3100-treated mice at week 4. CXCR4(+) cells were mobilized for just 1 day in both groups, but the mobilization of sca1(+)/flk1(+) cells endured for 7 days in AMD3100-treated mice compared with just 1 day in the saline-treated mice. AMD3100 upregulated BM levels of endothelial nitric oxide synthase (eNOS) and 2 targets of eNOS signaling, matrix metalloproteinase-9 and soluble Kit ligand. Furthermore, the loss of BM eNOS expression abolished the benefit of AMD3100 on sca1(+)/flk1(+) cell mobilization without altering the mobilization of CXCR4(+) cells, and the cardioprotective effects of AMD3100 were retained in eNOS-knockout mice that had been transplanted with BM from wild-type mice but not in wild-type mice with eNOS-knockout BM. CONCLUSIONS: AMD3100 prolongs BM progenitor mobilization and improves recovery from ischemia/reperfusion injury, and these benefits appear to occur through a previously unidentified link between AMD3100 and BM eNOS expression.


Asunto(s)
Compuestos Heterocíclicos/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Animales , Bencilaminas , Trasplante de Médula Ósea , Cardiotónicos/farmacología , Ciclamas , Modelos Animales de Enfermedad , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Recuperación de la Función/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
5.
Circ Res ; 111(3): 312-21, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22581926

RESUMEN

RATIONALE: Ischemic cardiovascular disease represents one of the largest epidemics currently facing the aging population. Current literature has illustrated the efficacy of autologous, stem cell therapies as novel strategies for treating these disorders. The CD34+ hematopoetic stem cell has shown significant promise in addressing myocardial ischemia by promoting angiogenesis that helps preserve the functionality of ischemic myocardium. Unfortunately, both viability and angiogenic quality of autologous CD34+ cells decline with advanced age and diminished cardiovascular health. OBJECTIVE: To offset age- and health-related angiogenic declines in CD34+ cells, we explored whether the therapeutic efficacy of human CD34+ cells could be enhanced by augmenting their secretion of the known angiogenic factor, sonic hedgehog (Shh). METHODS AND RESULTS: When injected into the border zone of mice after acute myocardial infarction, Shh-modified CD34+ cells (CD34(Shh)) protected against ventricular dilation and cardiac functional declines associated with acute myocardial infarction. Treatment with CD34(Shh) also reduced infarct size and increased border zone capillary density compared with unmodified CD34 cells or cells transfected with the empty vector. CD34(Shh) primarily store and secrete Shh protein in exosomes and this storage process appears to be cell-type specific. In vitro analysis of exosomes derived from CD34(Shh) revealed that (1) exosomes transfer Shh protein to other cell types, and (2) exosomal transfer of functional Shh elicits induction of the canonical Shh signaling pathway in recipient cells. CONCLUSIONS: Exosome-mediated delivery of Shh to ischemic myocardium represents a major mechanism explaining the observed preservation of cardiac function in mice treated with CD34(Shh) cells.


Asunto(s)
Antígenos CD34/administración & dosificación , Proteínas Hedgehog/administración & dosificación , Trasplante de Células Madre Hematopoyéticas/métodos , Infarto del Miocardio/cirugía , Animales , Antígenos CD34/uso terapéutico , Células Cultivadas , Proteínas Hedgehog/uso terapéutico , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Infarto del Miocardio/fisiopatología , Células 3T3 NIH , Disfunción Ventricular/fisiopatología , Disfunción Ventricular/cirugía
6.
Angiogenesis ; 16(1): 45-58, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22941227

RESUMEN

Neural stem cells (NSCs) differentiate into endothelial cells (ECs) and neuronal cells. Estradiol (E2) is known to exhibit proangiogenic effects on ischemic tissues via EC activation. Therefore, we hypothesized that E2 can promote the therapeutic potential of NSC transplantation for injured nerve repair via the differentiation of NSCs into ECs during neovascularization. NSCs isolated from newborn mouse brains were transplanted into injured sciatic nerves with (NSC/E2 group) or without E2-conjugated gelatin hydrogel (E2 group). The NSC/E2 group exhibited the greatest recovery in motor nerve conduction velocity, voltage amplitude, and exercise tolerance. Histological analyses revealed increased intraneural vascularity and blood perfusion as well as striking NSC recruitment to the neovasculature in the injured nerves in the NSC/E2 group. In vitro, E2 enhanced the NSC migration and proliferation inhibiting apoptosis. Fluorescence-activated cell sorting analysis also revealed that E2 significantly increased the percentage of CD31 in NSCs, and the effect of E2 was completely neutralized by the estrogen receptor antagonist ICI. The combination of E2 administration and NSC transplantation cooperatively improved the functional recovery of injured peripheral nerves, at least in part, via E2-associated NSC differentiation into ECs. These findings provide a novel mechanistic insight into both NSC biology and the biological effects of endogenous E2.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Células Endoteliales/citología , Estradiol/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Células-Madre Neurales/citología , Traumatismos de los Nervios Periféricos/terapia , Animales , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Células Endoteliales/efectos de los fármacos , Estradiol/sangre , Estradiol/uso terapéutico , Femenino , Ácido Láctico/administración & dosificación , Ácido Láctico/farmacología , Ratones , Ratones Endogámicos C57BL , Compresión Nerviosa , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/trasplante , Traumatismos de los Nervios Periféricos/sangre , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Traumatismos de los Nervios Periféricos/fisiopatología , Ácido Poliglicólico/administración & dosificación , Ácido Poliglicólico/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Recuperación de la Función/efectos de los fármacos , Nervio Ciático/efectos de los fármacos , Nervio Ciático/lesiones , Nervio Ciático/patología , Nervio Ciático/fisiopatología , Trasplante de Células Madre
7.
Circ Res ; 109(7): 724-8, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21835908

RESUMEN

RATIONALE: Transplantation of human CD34(+) stem cells to ischemic tissues has been associated with reduced angina, improved exercise time, and reduced amputation rates in phase 2 clinical trials and has been shown to induce neovascularization in preclinical models. Previous studies have suggested that paracrine factors secreted by these proangiogenic cells are responsible, at least in part, for the angiogenic effects induced by CD34(+) cell transplantation. OBJECTIVE: Our objective was to investigate the mechanism of CD34(+) stem cell-induced proangiogenic paracrine effects and to examine if exosomes, a component of paracrine secretion, are involved. METHODS AND RESULTS: Exosomes collected from the conditioned media of mobilized human CD34(+) cells had the characteristic size (40 to 90 nm; determined by dynamic light scattering), cup-shaped morphology (electron microscopy), expressed exosome-marker proteins CD63, phosphatidylserine (flow cytometry) and TSG101 (immunoblotting), besides expressing CD34(+) cell lineage marker protein, CD34. In vitro, CD34(+) exosomes replicated the angiogenic activity of CD34(+) cells by increasing endothelial cell viability, proliferation, and tube formation on Matrigel. In vivo, the CD34(+) exosomes stimulated angiogenesis in Matrigel plug and corneal assays. Interestingly, exosomes from CD34(+) cells but not from CD34(+) cell-depleted mononuclear cells had angiogenic activity. CONCLUSIONS: Our data demonstrate that human CD34(+) cells secrete exosomes that have independent angiogenic activity both in vitro and in vivo. CD34(+) exosomes may represent a significant component of the paracrine effect of progenitor cell transplantation for therapeutic angiogenesis.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Antígenos CD34/metabolismo , Neovascularización de la Córnea/metabolismo , Células Endoteliales/metabolismo , Exosomas/metabolismo , Neovascularización Fisiológica , Comunicación Paracrina , Células Madre/metabolismo , Animales , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Neovascularización de la Córnea/inmunología , Medios de Cultivo Condicionados/metabolismo , Proteínas de Unión al ADN/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Exosomas/inmunología , Exosomas/ultraestructura , Citometría de Flujo , Humanos , Luz , Masculino , Ratones , Ratones Desnudos , Microscopía Electrónica de Transmisión , Tamaño de la Partícula , Fosfatidilserinas/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Dispersión de Radiación , Células Madre/inmunología , Células Madre/ultraestructura , Tetraspanina 30 , Factores de Transcripción/metabolismo
8.
Proc Natl Acad Sci U S A ; 107(24): 11008-13, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20534467

RESUMEN

We hypothesized that a small molecule CXCR4 antagonist, AMD3100 (AMD), could augment the mobilization of bone marrow (BM)-derived endothelial progenitor cells (EPCs), thereby enhancing neovascularization and functional recovery after myocardial infarction. Single-dose AMD injection administered after the onset of myocardial infarction increased circulating EPC counts and myocardial vascularity, reduced fibrosis, and improved cardiac function and survival. In mice transplanted with traceable BM cells, AMD increased BM-derived cell incorporation in the ischemic border zone. In contrast, continuous infusion of AMD, although increasing EPCs in the circulation, worsened outcome by blocking EPC incorporation. In addition to its effects as a CXCR4 antagonist, AMD also up-regulated VEGF and matrix metalloproteinase 9 (MMP-9) expression, and the benefits of AMD were not observed in the absence of MMP-9 expression in the BM. These findings suggest that AMD3100 preserves cardiac function after myocardial infarction by enhancing BM-EPC-mediated neovascularization, and that these benefits require MMP-9 expression in the BM, but not in the ischemic region. Our results indicate that AMD3100 could be a potentially useful therapy for the treatment of myocardial infarction.


Asunto(s)
Trasplante de Médula Ósea , Infarto del Miocardio/terapia , Receptores CXCR4/antagonistas & inhibidores , Animales , Secuencia de Bases , Bencilaminas , Recuento de Células Sanguíneas , Capilares/efectos de los fármacos , Capilares/patología , Ciclamas , Cartilla de ADN/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Femenino , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Compuestos Heterocíclicos/farmacología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Neovascularización Fisiológica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Lab Invest ; 92(4): 532-42, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22330336

RESUMEN

Both estradiol (E2) and Sonic Hedgehog (Shh) contribute to angiogenesis and nerve regeneration. Here, we investigated whether E2 improves the recovery of injured nerves by downregulating the Shh inhibitor hedgehog-interacting protein (HIP) and increasing Shh-induced angiogenesis. Mice were treated with local injections of E2 or placebo one week before nerve-crush injury; 28 days after injury, nerve conduction velocity, exercise duration, and vascularity were significantly greater in E2-treated mice than in placebo-treated mice. E2 treatment was also associated with higher mRNA levels of Shh, the Shh receptor Patched-1, and the Shh transcriptional target Gli1, but with lower levels of HIP. The E2-induced enhancement of nerve vascularity was abolished by the Shh inhibitor cyclopamine, and the effect of E2 treatment on Shh, Gli1, and HIP mRNA expression was abolished by the E2 inhibitor ICI. Gli-luciferase activity in human umbilical-vein endothelial cells (HUVECs) increased more after treatment with E2 and Shh than after treatment with E2 alone, and E2 treatment reduced HIP expression in HUVECs and Schwann cells without altering Shh expression. Collectively, these findings suggest that E2 improves nerve recovery, at least in part, by reducing HIP expression, which subsequently leads to an increase in Shh signaling and Shh-induced angiogenesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Estradiol/metabolismo , Proteínas Hedgehog/metabolismo , Glicoproteínas de Membrana/metabolismo , Neovascularización Fisiológica , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/metabolismo , Animales , Regulación hacia Abajo , Estradiol/administración & dosificación , Femenino , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores Patched , Receptor Patched-1 , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Receptores de Superficie Celular/metabolismo , Recuperación de la Función , Transducción de Señal , Proteína con Dedos de Zinc GLI1
10.
J Exp Med ; 203(1): 153-63, 2006 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-16401693

RESUMEN

The cell surface receptor alpha4 integrin plays a critical role in the homing, engraftment, and maintenance of hematopoietic progenitor cells (HPCs) in the bone marrow (BM). Down-regulation or functional blockade of alpha4 integrin or its ligand vascular cell adhesion molecule-1 mobilizes long-term HPCs. We investigated the role of alpha4 integrin in the mobilization and homing of BM endothelial progenitor cells (EPCs). EPCs with endothelial colony-forming activity in the BM are exclusively alpha4 integrin-expressing cells. In vivo, a single dose of anti-alpha4 integrin antibody resulted in increased circulating EPC counts for 3 d. In hindlimb ischemia and myocardial infarction, systemically administered anti-alpha4 integrin antibody increased recruitment and incorporation of BM EPCs in newly formed vasculature and improved functional blood flow recovery and tissue preservation. Interestingly, BM EPCs that had been preblocked with anti-alpha4 integrin ex vivo or collected from alpha4 integrin-deficient mice incorporated as well as control cells into the neovasculature in ischemic sites, suggesting that alpha4 integrin may be dispensable or play a redundant role in EPC homing to ischemic tissue. These data indicate that functional disruption of alpha4 integrin may represent a potential angiogenic therapy for ischemic disease by increasing the available circulating supply of EPCs.


Asunto(s)
Integrina alfa4/metabolismo , Isquemia Miocárdica/fisiopatología , Neovascularización Fisiológica , Células Madre/fisiología , Animales , Médula Ósea , Movimiento Celular , Células Endoteliales , Integrina alfa4/genética , Masculino , Ratones , Ratones Noqueados
11.
Circ Res ; 107(9): 1083-93, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20847314

RESUMEN

RATIONALE: The mobilization of bone marrow (BM) progenitor cells (PCs) is largely governed by interactions between stromal cell-derived factor (SDF)-1 and CXC chemokine receptor (CXCR)4. Ischemic injury disrupts the SDF-1-CXCR4 interaction and releases BM PCs into the peripheral circulation, where the mobilized cells are recruited to the injured tissue and contribute to vessel growth. BM PCs can also be mobilized by the pharmacological CXCR4 antagonist AMD3100, but the other components of the SDF-1-CXCR4 signaling pathway are largely unknown. c-kit, a membrane-bound tyrosine kinase and the receptor for stem cell factor, has also been shown to play a critical role in BM PC mobilization and ischemic tissue repair. OBJECTIVE: To investigate the functional interaction between SDF-1-CXCR4 signaling and c-kit activity in BM PC mobilization. METHODS AND RESULTS: AMD3100 administration failed to mobilize BM PCs in mice defective in c-kit kinase activity or in mice transplanted with BM cells that expressed a constitutively active c-kit mutant. Furthermore, BM levels of phosphorylated (phospho)-c-kit declined after AMD3100 administration and after CXCR4 deletion. In cells adhering to culture plates coated with vascular cell adhesion molecule 1, SDF-1 and stem cell factor increased phospho-c-kit levels, and AMD3100 treatment suppressed SDF-1-induced, but not SCF-induced, c-kit phosphorylation. SDF-1-induced c-kit phosphorylation also required the activation of Src nonreceptor tyrosine kinase: pretreatment of cells with a selective Src inhibitor blocked both c-kit phosphorylation and the interaction between c-kit and phospho-Src. CONCLUSIONS: These findings indicate that the regulation of BM PC trafficking by SDF-1 and CXCR4 is dependent on Src-mediated c-kit phosphorylation.


Asunto(s)
Células de la Médula Ósea/fisiología , Movimiento Celular/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores CXCR4/fisiología , Células Madre/fisiología , Animales , Bencilaminas , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/enzimología , Línea Celular , Movimiento Celular/genética , Quimiocina CXCL12/fisiología , Ciclamas , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Compuestos Heterocíclicos/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-kit/deficiencia , Proteínas Proto-Oncogénicas c-kit/genética , Receptores CXCR4/deficiencia , Receptores CXCR4/genética , Células Madre/efectos de los fármacos , Células Madre/enzimología , Familia-src Quinasas/fisiología
12.
Nat Med ; 11(11): 1197-204, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16244652

RESUMEN

Sonic hedgehog (Shh) is a crucial regulator of organ development during embryogenesis. We investigated whether intramyocardial gene transfer of naked DNA encoding human Shh (phShh) could promote a favorable effect on recovery from acute and chronic myocardial ischemia in adult animals, not only by promoting neovascularization, but by broader effects, consistent with the role of this morphogen in embryogenesis. After Shh gene transfer, the hedgehog pathway was upregulated in mammalian fibroblasts and cardiomyocytes. This resulted in preservation of left ventricular function in both acute and chronic myocardial ischemia by enhanced neovascularization, and reduced fibrosis and cardiac apoptosis. Shh gene transfer also enhanced the contribution of bone marrow-derived endothelial progenitor cells to myocardial neovascularization. These data suggest that Shh gene therapy may have considerable therapeutic potential in individuals with acute and chronic myocardial ischemia by triggering expression of multiple trophic factors and engendering tissue repair in the adult heart.


Asunto(s)
Terapia Genética , Corazón/embriología , Miocardio/metabolismo , Transducción de Señal , Transactivadores/uso terapéutico , Enfermedad Aguda , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Enfermedad Crónica , Modelos Animales de Enfermedad , Ecocardiografía , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog , Humanos , Ratones , Ratones Mutantes , Isquemia Miocárdica/etiología , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/terapia , Miocardio/citología , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Función Ventricular Izquierda/fisiología
13.
Circulation ; 121(9): 1104-12, 2010 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-20176991

RESUMEN

BACKGROUND: Little is known about the role of endothelial progenitor cells (EPCs) in atherosclerosis. Accordingly, we performed a series of assessments with hypercholesterolemic (apolipoprotein E-null [ApoE(-/-)]) and wild-type (WT) mice to evaluate how cholesterol influences reendothelialization, atherosclerosis, and EPC function after arterial injury. METHODS AND RESULTS: Unexpectedly, reendothelialization (assessed by resistance to Evans blue staining) and circulating EPC counts (EPC culture assay) were greater in ApoE(-/-) mice than in WT mice, and transplantation of ApoE(-/-) bone marrow in WT mice accelerated endothelial recovery and increased recruitment of bone marrow-derived EPCs to the neoendothelium. Cholesterol concentration-dependently promoted the proliferation (MTS assay) of both ApoE(-/-) and WT EPCs, and the concentration dependence of EPC adhesion (to vitronectin-, collagen type I-, fibronectin-, and laminin-coated plates), migration (modified Boyden chamber assay), and antiapoptotic (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling stain) activity was biphasic. Cholesterol enhanced the messenger RNA expression (quantitative, real-time reverse-transcription polymerase chain reaction) of vascular endothelial growth factor and inhibited Notch1 messenger RNA expression in both ApoE(-/-) and WT EPCs, whereas endothelial nitric oxide synthase messenger RNA expression increased in ApoE(-/-) EPCs and declined in WT EPCs after cholesterol exposure. EPC activity was greater in Notch1(+/-) EPCs than in WT EPCs, and transplantation of Notch1(+/-) bone marrow accelerated endothelial recovery after arterial injury in WT mice. CONCLUSIONS: The results presented here provide novel insights into the role of EPCs during atherosclerosis and suggest that cholesterol and Notch1 may be involved in the regulation of EPC activity.


Asunto(s)
Traumatismos de las Arterias Carótidas/patología , Células Endoteliales/patología , Hipercolesterolemia/patología , Células Madre Mesenquimatosas/patología , Receptor Notch1/fisiología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/patología , Trasplante de Médula Ósea , Traumatismos de las Arterias Carótidas/complicaciones , Movimiento Celular , Colesterol/sangre , Regulación de la Expresión Génica , Genotipo , Hipercolesterolemia/complicaciones , Hipercolesterolemia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Óxido Nítrico Sintasa de Tipo III/genética , ARN Mensajero/biosíntesis , Quimera por Radiación , Receptor Notch1/biosíntesis , Receptor Notch1/deficiencia , Receptor Notch1/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
14.
FASEB J ; 24(7): 2484-94, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20219984

RESUMEN

Prolonged inflammatory response is associated with left ventricular (LV) dysfunction and adverse remodeling following myocardial infarction (MI). IL-10 inhibits inflammation by suppressing HuR-mediated mRNA stabilization of proinflammatory cytokines. Here we report that following MI, IL-10(-/-) mice showed exaggerated LV dysfunction, fibrosis, and cardiomyocyte apoptosis. Short-hairpin RNA (shRNA)-mediated knockdown of HuR in the myocardium significantly reversed MI-induced LV dysfunctions and LV remodeling. HuR knockdown significantly reduced MI-induced cardiomyocyte apoptosis concomitant with reduced p53 expression. Moreover, HuR knockdown significantly reduced infarct size and fibrosis area, which in turn was associated with decreased TGF-beta expression. In vitro, stable knockdown of HuR in mouse macrophage cell line RAW 264.7 corroborated in vivo data and revealed reduced mRNA expression of TNF-alpha, TGF-beta, and p53 following LPS challenge, which was associated with a marked reduction in the mRNA stability of these genes. Taken together, our studies suggest that HuR is a direct target of IL-10, and HuR knockdown mimics anti-inflammatory effects of IL-10.


Asunto(s)
Antígenos de Superficie/genética , Inflamación/prevención & control , Interleucina-10/deficiencia , Infarto del Miocardio/patología , Miocardio/patología , Proteínas de Unión al ARN/genética , Disfunción Ventricular Izquierda/prevención & control , Animales , Antígenos de Superficie/fisiología , Línea Celular , Proteínas ELAV , Proteína 1 Similar a ELAV , Inflamación/etiología , Interleucina-10/fisiología , Macrófagos , Ratones , Ratones Noqueados , Miocardio/metabolismo , Estabilidad del ARN , ARN Interferente Pequeño/farmacología , Proteínas de Unión al ARN/fisiología
15.
Circ Res ; 105(8): 818-26, 2009 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-19729595

RESUMEN

RATIONALE: The Gli transcription factors are mediators of Hedgehog (Hh) signaling and have been shown to play critical roles during embryogenesis. Previously, we have demonstrated that the Hh pathway is reactivated by ischemia in adult mammals, and that this pathway can be stimulated for therapeutic benefit; however, the specific roles of the Gli transcription factors during ischemia-induced Hh signaling have not been elucidated. OBJECTIVE: To investigate the role of Gli3 in ischemic tissue repair. METHODS AND RESULTS: Gli3-haploinsufficient (Gli3(+/-)) mice and their wild-type littermates were physiologically similar in the absence of ischemia; however, histological assessments of capillary density and echocardiographic measurements of left ventricular ejection fractions were reduced in Gli3(+/-) mice compared to wild-type mice after surgically induced myocardial infarction, and fibrosis was increased. Gli3-deficient mice also displayed reduced capillary density after induction of hindlimb ischemia and an impaired angiogenic response to vascular endothelial growth factor in the corneal angiogenesis model. In endothelial cells, adenovirus-mediated overexpression of Gli3 promoted migration (modified Boyden chamber), small interfering RNA-mediated downregulation of Gli3 delayed tube formation (Matrigel), and Western analyses identified increases in Akt phosphorylation, extracellular signal-regulated kinase (ERK)1/2 activation, and c-Fos expression; however, promoter-reporter assays indicated that Gli3 overexpression does not modulate Gli-dependent transcription. Furthermore, the induction of endothelial cell migration by Gli3 was dependent on Akt and ERK1/2 activation. CONCLUSIONS: Collectively, these observations indicate that Gli3 contributes to vessel growth under both ischemic and nonischemic conditions and provide the first evidence that Gli3 regulates angiogenesis and endothelial cell activity in adult mammals.


Asunto(s)
Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Infarto del Miocardio/metabolismo , Neovascularización Fisiológica , Proteínas del Tejido Nervioso/metabolismo , Regeneración , Transducción de Señal , Animales , Línea Celular , Movimiento Celular/genética , Femenino , Fibrosis , Regulación de la Expresión Génica/genética , Genes fos/genética , Proteínas Hedgehog/genética , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Transgénicos , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Proteínas del Tejido Nervioso/genética , Proteína Gli3 con Dedos de Zinc
16.
J Mol Cell Cardiol ; 49(3): 490-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20478312

RESUMEN

The morphogen Sonic hedgehog (Shh) promotes neovascularization in adults by inducing pro-angiogenic cytokine expression in fibroblasts; however, the direct effects of Shh on endothelial cell (EC) function during angiogenesis are unknown. Our findings indicate that Shh promotes capillary morphogenesis (tube length on Matrigel increased to 271+/-50% of the length in untreated cells, p=0.00003), induces EC migration (modified Boyden chamber assay, 191+/-35% of migration in untreated cells, p=0.00009), and increases EC expression of matrix metalloproteinase 9 (MMP-9) and osteopontin (OPN) mRNA (real-time RT-PCR), which are essential for Shh-induced angiogenesis both in vitro and in vivo. Shh activity in ECs is mediated by Rho, rather than through the "classic" Shh signaling pathway, which involves the Gli transcription factors. The Rho dependence of Shh-induced EC angiogenic activity was documented both in vitro, with dominant-negative RhoA and Rho kinase (ROCK) constructs, and in vivo, with the ROCK inhibitor Y27632 in the mouse corneal angiogenesis model. Finally, experiments performed in MMP-9- and OPN-knockout mice confirmed the roles of the ROCK downstream targets MMP-9 and OPN in Shh-induced angiogenesis. Collectively, our results identify a "nonclassical" pathway by which Shh directly modulates EC phenotype and angiogenic activity.


Asunto(s)
Aorta/metabolismo , Neovascularización de la Córnea/metabolismo , Endotelio Vascular/metabolismo , Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Neovascularización Fisiológica/fisiología , Quinasas Asociadas a rho/metabolismo , Animales , Aorta/citología , Aorta/efectos de los fármacos , Apoptosis , Western Blotting , Bovinos , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Neovascularización de la Córnea/patología , Vasos Coronarios/citología , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Proteínas Hedgehog/genética , Humanos , Factores de Transcripción de Tipo Kruppel/fisiología , Metaloproteinasa 9 de la Matriz/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Morfogénesis , Proteínas del Tejido Nervioso/fisiología , Osteopontina/fisiología , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína Gli3 con Dedos de Zinc , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética
17.
Circulation ; 120(13): 1213-21, 2009 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-19752322

RESUMEN

BACKGROUND: Recent studies have identified a polymorphism in the endothelin-converting enzyme (ECE)-1b promoter (-338C/A) that is strongly associated with hypertension in women. The polymorphism is located in a consensus binding sequence for the E2F family of transcription factors. E2F proteins are crucially involved in cell-cycle regulation, but their roles in cardiovascular function are poorly understood. Here, we investigated the potential role of E2F2 in blood pressure regulation. METHODS AND RESULTS: Tail-cuff measurements of systolic and diastolic blood pressures were significantly higher in E2F2-null (E2F2(-/-)) mice than in their wild-type littermates, and in ex vivo ring assays, aortas from the E2F2(-/-) mice exhibited significantly greater contractility in response to big endothelin-1. Big endothelin-1 is activated by ECE-1, and mRNA levels of ECE-1b, the repressive ECE-1 isoform, were significantly lower in E2F2(-/-) mice than in wild-type mice. In endothelial cells, chromatin immunoprecipitation assays confirmed that E2F2 binds the ECE-1b promoter, and promoter-reporter assays indicated that E2F2 activates ECE-1b transcription. Furthermore, loss or downregulation of E2F2 led to a decline in ECE-1b levels, to higher levels of the membranous ECE-1 isoforms (ie, ECE-1a, -1c, and -1d), and to deregulated ECE-1 activity. Finally, Sam68 coimmunoprecipitated with E2F2, occupied the ECE-1b promoter (chromatin immunoprecipitation), and repressed E2F2-mediated ECE-1b promoter activity (promoter-reporter assays). CONCLUSIONS: Our results identify a cell-cycle-independent mechanism by which E2F2 regulates endothelial function, arterial contractility, and blood pressure.


Asunto(s)
Presión Sanguínea/fisiología , Factor de Transcripción E2F2/genética , Factor de Transcripción E2F2/metabolismo , Activación Transcripcional/fisiología , Vasoconstricción/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Aorta/fisiología , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Ciclo Celular/fisiología , Membrana Celular/metabolismo , Regulación hacia Abajo/fisiología , Endotelina-1/farmacología , Enzimas Convertidoras de Endotelina , Femenino , Masculino , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Ratones , Ratones Mutantes , Regiones Promotoras Genéticas/fisiología , Proteínas de Unión al ARN/metabolismo , Vasoconstricción/efectos de los fármacos
18.
Circ Res ; 102(11): e107-17, 2008 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-18483406

RESUMEN

The oocyte-independent source for the generation of pluripotent stem cells is among the ultimate goals in regenerative medicine. We report that on exposure to mouse embryonic stem cell (mESC) extracts, reversibly permeabilized NIH3T3 cells undergo dedifferentiation followed by stimulus-induced redifferentiation into multiple lineage cell types. Genome-wide expression profiling revealed significant differences between NIH3T3 control and ESC extract-treated NIH3T3 cells including the reactivation of ESC-specific transcripts. Epigenetically, ESC extracts induced CpG demethylation of Oct4 promoter, hyperacetylation of histones 3 and 4, and decreased lysine 9 (K-9) dimethylation of histone 3. In mouse models of surgically induced hindlimb ischemia or acute myocardial infarction transplantation of reprogrammed NIH3T3 cells significantly improved postinjury physiological functions and showed anatomic evidence of engraftment and transdifferentiation into skeletal muscle, endothelial cell, and cardiomyocytes. These data provide evidence for the generation of functional multipotent stem-like cells from terminally differentiated somatic cells without the introduction of retroviral mediated transgenes or ESC fusion.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Extractos Celulares/farmacología , Fibroblastos/efectos de los fármacos , Isquemia/terapia , Células Madre Multipotentes/citología , Infarto del Miocardio/terapia , Animales , Diferenciación Celular/genética , Extractos Celulares/química , Línea Celular , Linaje de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Células Madre Embrionarias/química , Fibroblastos/citología , Fibroblastos/trasplante , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Miembro Posterior/irrigación sanguínea , Miembro Posterior/fisiopatología , Isquemia/patología , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/trasplante , Infarto del Miocardio/patología , Células 3T3 NIH , Ingeniería de Tejidos
19.
Circulation ; 115(6): 752-62, 2007 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-17261656

RESUMEN

BACKGROUND: Aging is a risk factor for coronary and peripheral artery disease. Tumor necrosis factor-alpha (TNF-alpha), a proinflammatory cytokine, is expressed in ischemic tissue and is known to modulate angiogenesis. Little is known about the role of TNF-alpha receptors (TNFR1/p55 and TNFR2/p75) in angiogenic signaling. METHODS AND RESULTS: We studied neovascularization in the hindlimb ischemia model in young and old TNFR2/p75 knockout (p75KO) and wild-type age-matched controls. Between days 7 to 10 after hindlimb surgery, 100% of old p75KOs experienced autoamputation of the operated limbs, whereas none of the age-matched wild-type mice exhibited hindlimb necrosis. Poor blood flow recovery in p75KO mice was associated with increased endothelial cell apoptosis, decreased capillary density, and significant reductions in the expression of vascular endothelial growth factor and basic fibroblast growth factor-2 mRNA transcripts in ischemic tissue and in circulating endothelial progenitor cells. The number of circulating bone marrow-derived endothelial progenitor cells was significantly reduced in p75KO mice. Transplantation of wild-type bone marrow mononuclear cells into irradiated old p75KO mice 1 month before hindlimb surgery prevented limb loss. CONCLUSIONS: Our present study suggests that ischemia-induced endothelial progenitor cell-mediated neovascularization is dependent, at least in part, on p75 TNF receptor expressed in bone marrow-derived cells. Specifically, endothelial cell/endothelial progenitor cell survival, vascular endothelial growth factor expression, endothelial progenitor cell mobilization from bone marrow, endothelial progenitor cell differentiation, and ultimately ischemia-induced collateral vessel development are dependent on signaling through TNFR2/p75. Furthermore, because TNFR2/p75 becomes an age-related limiting factor in postischemic recovery, it may be a potential gene target for therapeutic interventions in adult vascular diseases.


Asunto(s)
Isquemia/fisiopatología , Neovascularización Fisiológica , Receptores Tipo II del Factor de Necrosis Tumoral/fisiología , Envejecimiento/fisiología , Animales , Apoptosis , Trasplante de Médula Ósea , Células Cultivadas , Células Endoteliales/patología , Miembro Posterior/irrigación sanguínea , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Transducción de Señal , Células Madre/fisiología , Factor A de Crecimiento Endotelial Vascular/genética
20.
Circulation ; 115(25): 3165-72, 2007 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-17562958

RESUMEN

BACKGROUND: A growing population of patients with coronary artery disease experiences angina that is not amenable to revascularization and is refractory to medical therapy. Preclinical studies have indicated that human CD34+ stem cells induce neovascularization in ischemic myocardium, which enhances perfusion and function. METHODS AND RESULTS: Twenty-four patients (19 men and 5 women aged 48 to 84 years) with Canadian Cardiovascular Society class 3 or 4 angina who were undergoing optimal medical treatment and who were not candidates for mechanical revascularization were enrolled in a double-blind, randomized (3:1), placebo-controlled dose-escalating study. Patients received granulocyte colony-stimulating factor 5 microg x kg(-1) x d(-1) for 5 days with leukapheresis on the fifth day. Selection of CD34+ cells was performed with a Food and Drug Administration-approved device. Electromechanical mapping was performed to identify ischemic but viable regions of myocardium for injection of cells (versus saline). The total dose of cells was distributed in 10 intramyocardial, transendocardial injections. Patients were required to have an implantable cardioverter-defibrillator or to temporarily wear a LifeVest wearable defibrillator. No incidence was observed of myocardial infarction induced by mobilization or intramyocardial injection. The intramyocardial injection of cells or saline did not result in cardiac enzyme elevation, perforation, or pericardial effusion. No incidence of ventricular tachycardia or ventricular fibrillation occurred during the administration of granulocyte colony-stimulating factor or intramyocardial injections. One patient with a history of sudden cardiac death/ventricular tachycardia/ventricular fibrillation had catheter-induced ventricular tachycardia during mapping that required cardioversion. Serious adverse events were evenly distributed. Efficacy parameters including angina frequency, nitroglycerine usage, exercise time, and Canadian Cardiovascular Society class showed trends that favored CD34+ cell-treated patients versus control subjects given placebo. CONCLUSIONS: A randomized trial of intramyocardial injection of autologous CD34+ cells in patients with intractable angina was completed that provides evidence for feasibility, safety, and bioactivity. A larger phase IIb study is currently under way to further evaluate this therapy.


Asunto(s)
Angina de Pecho/cirugía , Trasplante de Células Madre de Sangre Periférica , Anciano , Anciano de 80 o más Años , Angina de Pecho/inducido químicamente , Angina de Pecho/diagnóstico por imagen , Angina de Pecho/tratamiento farmacológico , Fármacos Cardiovasculares/uso terapéutico , Recuento de Células , Terapia Combinada , Método Doble Ciego , Cardioversión Eléctrica , Electrocardiografía Ambulatoria , Tolerancia al Ejercicio , Femenino , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Inyecciones , Masculino , Persona de Mediana Edad , Miocardio , Trasplante de Células Madre de Sangre Periférica/métodos , Calidad de Vida , Tomografía Computarizada de Emisión de Fotón Único , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA