Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Circ Res ; 106(3): 479-94, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20167944

RESUMO

Myocardial regeneration using stem and progenitor cell transplantation in the injured heart has recently become a major goal in the treatment of cardiac disease. Experimental studies and clinical applications have generally been encouraging, although the functional benefits that have been attained clinically are modest and inconsistent. Low cell retention and engraftment after myocardial delivery is a key factor limiting the successful application of cell therapy, irrespective of the type of cell or the delivery method. To improve engraftment, accurate methods for tracking cell fate and quantifying cell survival need to be applied. Several laboratory techniques (histological methods, real-time quantitative polymerase chain reaction, radiolabeling) have provided invaluable information about cell engraftment. In vivo imaging (nuclear medicine modalities, bioluminescence, and MRI) has the potential to provide quantitative information noninvasively, enabling longitudinal assessment of cell fate. In the present review, we present several available methods for assessing cell engraftment, and we critically discuss their strengths and limitations. In addition to providing insights about the mechanisms mediating cell loss after transplantation, these methods can evaluate techniques for augmenting engraftment, such as tissue engineering approaches, preconditioning, and genetic modification, allowing optimization of cell therapies.


Assuntos
Imagem Molecular/métodos , Infarto do Miocárdio/cirurgia , Transplante de Células-Tronco , Animais , Linhagem da Célula , Sobrevivência Celular , Genes Reporter , Sobrevivência de Enxerto , Humanos , Hibridização in Situ Fluorescente , Camundongos , Camundongos SCID , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Tomografia por Emissão de Pósitrons , Pontos Quânticos , Ratos , Suínos , Tomografia Computadorizada de Emissão de Fóton Único , Transplante Heterólogo
2.
Circ Res ; 106(5): 971-80, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20110532

RESUMO

RATIONALE: Multiple biological mechanisms contribute to the efficacy of cardiac cell therapy. Most prominent among these are direct heart muscle and blood vessel regeneration from transplanted cells, as opposed to paracrine enhancement of tissue preservation and/or recruitment of endogenous repair. OBJECTIVE: Human cardiac progenitor cells, cultured as cardiospheres (CSps) or as CSp-derived cells (CDCs), have been shown to be capable of direct cardiac regeneration in vivo. Here we characterized paracrine effects in CDC transplantation and investigated their relative importance versus direct differentiation of surviving transplanted cells. METHODS AND RESULTS: In vitro, many growth factors were found in media conditioned by human adult CSps and CDCs; CDC-conditioned media exerted antiapoptotic effects on neonatal rat ventricular myocytes, and proangiogenic effects on human umbilical vein endothelial cells. In vivo, human CDCs secreted vascular endothelial growth factor, hepatocyte growth factor, and insulin-like growth factor 1 when transplanted into the same SCID mouse model of acute myocardial infarction where they were previously shown to improve function and to produce tissue regeneration. Injection of CDCs in the peri-infarct zone increased the expression of Akt, decreased apoptotic rate and caspase 3 level, and increased capillary density, indicating overall higher tissue resilience. Based on the number of human-specific cells relative to overall increases in capillary density and myocardial viability, direct differentiation quantitatively accounted for 20% to 50% of the observed effects. CONCLUSIONS: Together with their spontaneous commitment to cardiac and angiogenic differentiation, transplanted CDCs serve as "role models," recruiting endogenous regeneration and improving tissue resistance to ischemic stress. The contribution of the role model effect rivals or exceeds that of direct regeneration.


Assuntos
Células Endoteliais/transplante , Infarto do Miocárdio/cirurgia , Miócitos Cardíacos/transplante , Comunicação Parácrina , Regeneração , Transplante de Células-Tronco , Animais , Animais Recém-Nascidos , Apoptose , Caspase 3/metabolismo , Diferenciação Celular , Linhagem da Célula , Sobrevivência Celular , Meios de Cultivo Condicionados/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos SCID , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Neovascularização Fisiológica , Análise Serial de Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Esferoides Celulares , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Stem Cells ; 28(11): 2088-98, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20882531

RESUMO

Cardiac stem cells (CSCs) are promising candidates for use in myocardial regenerative therapy. We test the hypothesis that growing cardiac-derived cells as three-dimensional cardiospheres may recapitulate a stem cell niche-like microenvironment, favoring cell survival and enhancing functional benefit after transplantation into the injured heart. CSCs and supporting cells from human endomyocardial biopsies were grown as cardiospheres and compared with cells cultured under traditional monolayer condition or dissociated from cardiospheres. Cardiospheres self-assembled into stem cell niche-like structures in vitro in suspension culture, while exhibiting greater proportions of c-kit(+) cells and upregulated expression of SOX2 and Nanog. Pathway-focused polymerase chain reaction (PCR) array, quantitative real-time PCR, and immunostaining revealed enhanced expression of stem cell-relevant factors and adhesion/extracellular-matrix molecules (ECM) in cardiospheres including IGF-1, histone deacetylase 2 (HDAC2), Tert, integrin-α(2), laminin-ß(1), and matrix metalloproteinases (MMPs). Implantation of cardiospheres in severe combined immunodeficiency (SCID) mouse hearts with acute infarction disproportionately improved cell engraftment and myocardial function, relative to monolayer-cultured cells. Dissociation of cardiospheres into single cells decreased the expression of ECM and adhesion molecules and undermined resistance to oxidative stress, negating the improved cell engraftment and functional benefit in vivo. Growth of cardiac-derived cells as cardiospheres mimics stem cell niche properties with enhanced "stemness" and expression of ECM and adhesion molecules. These changes underlie an increase in cell survival and more potent augmentation of global function following implantation into the infarcted heart.


Assuntos
Miocárdio/citologia , Células-Tronco/citologia , Animais , Western Blotting , Células Cultivadas , Ecocardiografia , Matriz Extracelular/metabolismo , Citometria de Fluxo , Histona Desacetilase 2/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Integrina alfa2/metabolismo , Laminina/metabolismo , Camundongos , Camundongos SCID , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/terapia , Miocárdio/metabolismo , Estresse Oxidativo/fisiologia , Reação em Cadeia da Polimerase , Transplante de Células-Tronco , Células-Tronco/metabolismo , Telomerase/metabolismo
4.
J Mol Cell Cardiol ; 49(2): 312-21, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20211627

RESUMO

The adult heart contains reservoirs of progenitor cells that express embryonic and stem cell-related antigens. While these antigenically-purified cells are promising candidates for autologous cell therapy, clinical application is hampered by their limited abundance and tedious isolation methods. Methods that involve an intermediate cardiosphere-forming step have proven successful and are being tested clinically, but it is unclear whether the cardiosphere step is necessary. Accordingly, we investigated the molecular profile and functional benefit of cells that spontaneously emigrate from cardiac tissue in primary culture. Adult Wistar-Kyoto rat hearts were minced, digested and cultured as separate anatomical regions. Loosely-adherent cells that surround the plated tissue were harvested weekly for a total of five harvests. Genetic lineage tracing demonstrated that a small proportion of the direct outgrowth from cardiac samples originates from myocardial cells. This outgrowth contains sub-populations of cells expressing embryonic (SSEA-1) and stem cell-related antigens (c-Kit, abcg2) that varied with time in culture but not with the cardiac chamber of origin. This direct outgrowth, and its expanded progeny, underwent marked in vitro angiogenic/cardiogenic differentiation and cytokine secretion (IGF-1, VGEF). In vivo effects included long-term functional benefits as gauged by MRI following cell injection in a rat model of myocardial infarction. Outgrowth cells afforded equivalent functional benefits to cardiosphere-derived cells, which require more processing steps to manufacture. These results provide the basis for a simplified and efficient process to generate autologous cardiac progenitor cells (and mesenchymal supporting cells) to augment clinically-relevant approaches for myocardial repair.


Assuntos
Separação Celular/métodos , Miocárdio/patologia , Células-Tronco/citologia , Indutores da Angiogênese/metabolismo , Animais , Biomarcadores/metabolismo , Biópsia , Cardiotônicos/metabolismo , Diferenciação Celular , Linhagem da Célula , Membrana Celular/metabolismo , Proliferação de Células , Citocinas/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Citometria de Fluxo , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/citologia , Neovascularização Fisiológica , Fenótipo , Ratos , Ratos Wistar , Transplante de Células-Tronco , Células-Tronco/metabolismo , Função Ventricular/fisiologia
5.
Circulation ; 120(12): 1075-83, 7 p following 1083, 2009 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-19738142

RESUMO

BACKGROUND: Cardiosphere-derived cells (CDCs) isolated from human endomyocardial biopsies reduce infarct size and improve cardiac function in mice. Safety and efficacy testing in large animals is necessary for clinical translation. METHODS AND RESULTS: Mesenchymal stem cells, which resemble CDCs in size and thrombogenicity, have been associated with infarction after intracoronary infusion. To maximize CDC engraftment while avoiding infarction, we optimized the infusion protocol in 19 healthy pigs. A modified cocktail of CDCs in calcium-free PBS, 100 U/mL of heparin, and 250 microg/mL of nitroglycerin eliminated infusion-related infarction. Subsequent infusion experiments in 17 pigs with postinfarct left ventricular dysfunction showed CDC doses > or =10(7) but <2.5 x 10(7) result in new myocardial tissue formation without infarction. In a pivotal randomized study, 7 infarcted pigs received 300,000 CDCs/kg (approximately 10(7) total) and 7 received placebo (vehicle alone). Cardiac magnetic resonance imaging 8 weeks later showed CDC treatment decreased relative infarct size (19.2% to 14.2% of left ventricle infarcted, P=0.01), whereas placebo did not (17.7% to 15.3%, P=0.22). End-diastolic volume increased in placebo, but not in CDC-treated animals. Hemodynamically, the rate of pressure change (dP/dt) maximum and dP/dt minimum were significantly better with CDC infusion. There was no difference between groups in the ability to induce ventricular tachycardia, nor was there any tumor or ectopic tissue formation. CONCLUSIONS: Intracoronary delivery of CDCs in a preclinical model of postinfarct left ventricular dysfunction results in formation of new cardiac tissue, reduces relative infarct size, attenuates adverse remodeling, and improves hemodynamics. The evidence of efficacy without obvious safety concerns at 8 weeks of follow-up motivates human studies in patients after myocardial infarction and in chronic ischemic cardiomyopathy.


Assuntos
Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Transplante de Células-Tronco , Animais , Biópsia , Diferenciação Celular , Separação Celular , Hemodinâmica , Humanos , Infarto do Miocárdio/fisiopatologia , Células-Tronco/fisiologia , Suínos , Trombose/etiologia , Transplante Autólogo
6.
J Am Soc Nephrol ; 20(4): 831-41, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19297559

RESUMO

Multipotent mesenchymal stromal cells from the bone marrow ameliorate acute kidney injury through a mechanism other than transdifferentiation into renal tissue. Stromal cells exert immunoregulatory effects on dendritic and T cells, both of which are important in the pathophysiology of immune-mediated kidney injury. We hypothesized that similar cells with immunoregulatory function exist within the adult kidney. We isolated murine kidney-derived cells with morphologic features, growth properties, and an immunophenotype characteristic of mesenchymal stromal cells. These cells lacked lineage markers and could be differentiated into mesodermal cell lineages, including osteocytes and adipocytes. Furthermore, these kidney-derived cells induced the generation of bone marrow-derived dendritic cells with significantly reduced MHC II expression, increased CD80 expression, increased IL-10 production and the inability to stimulate CD4+ T cell proliferation in allogeneic and nominal antigen-specific cultures. Experiments in mixed and transwell cultures demonstrated that the production of soluble immune modulators, such as IL-6, was responsible for these effects on dendritic cell differentiation and maturation. Contact-dependent mechanisms, however, inhibited mitogenic T cell proliferation. In summary, kidney-derived cells may suppress inflammation in the kidney in vivo; a better understanding of their biology could have therapeutic implications in a wide variety of immune-mediated kidney diseases.


Assuntos
Células Dendríticas/imunologia , Rim/fisiologia , Células Estromais/fisiologia , Linfócitos T/imunologia , Adipócitos/citologia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/fisiologia , Diferenciação Celular , Células Dendríticas/citologia , Humanos , Imunofenotipagem , Inflamação/fisiopatologia , Inflamação/prevenção & controle , Rim/imunologia , Ativação Linfocitária/imunologia , Camundongos , Osteócitos/citologia , Células Estromais/imunologia , Linfócitos T/citologia
7.
Neurology ; 92(8): e866-e878, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30674601

RESUMO

OBJECTIVE: To assess the feasibility, safety, and efficacy of intracoronary allogeneic cardiosphere-derived cells (CAP-1002) in patients with Duchenne muscular dystrophy (DMD). METHODS: The Halt Cardiomyopathy Progression (HOPE)-Duchenne trial is a phase I/II, randomized, controlled, open-label trial (NCT02485938). Patients with DMD >12 years old, with substantial myocardial fibrosis, were randomized (1:1) to usual care (control) or global intracoronary infusion of CAP-1002 (75 million cells). Participants were enrolled at 3 US medical centers between January and August 2016 and followed for 12 months. An independent Data and Safety Monitoring Board provided safety oversight. Cardiac function and structure were assessed by MRI, and analyzed by a blinded core laboratory. Skeletal muscle function was assessed by performance of the upper limb (PUL). RESULTS: Twenty-five eligible patients (mean age 17.8 years; 68% wheelchair-dependent) were randomized to CAP-1002 (n = 13) or control (n = 12). Incidence of treatment-emergent adverse events was similar between groups. Compared to baseline, MRI at 12 months revealed significant scar size reduction and improvement in inferior wall systolic thickening in CAP-1002 but not control patients. Mid-distal PUL improved at 12 months in 8 of 9 lower functioning CAP-1002 patients, and no controls (p = 0.007). CONCLUSIONS: Intracoronary CAP-1002 in DMD appears safe and demonstrates signals of efficacy on both cardiac and upper limb function for up to 12 months. Thus, future clinical research on CAP-1002 treatment of DMD cardiac and skeletal myopathies is warranted. CLASSIFICATION OF EVIDENCE: This phase I/II study provides Class II evidence that for patients with DMD, intracoronary CAP-1002 is feasible and appears safe and potentially effective.


Assuntos
Cardiomiopatias/terapia , Distrofia Muscular de Duchenne/terapia , Transplante de Células-Tronco/métodos , Atividades Cotidianas , Adolescente , Adulto , Células Alógenas , Cardiomiopatias/diagnóstico por imagem , Cardiomiopatias/etiologia , Cardiomiopatias/fisiopatologia , Terapia Baseada em Transplante de Células e Tecidos , Estudos de Viabilidade , Fibrose , Humanos , Imageamento por Ressonância Magnética , Masculino , Distrofia Muscular de Duchenne/complicações , Distrofia Muscular de Duchenne/fisiopatologia , Miocárdio/patologia , Qualidade de Vida , Espirometria , Transplante Homólogo , Extremidade Superior/fisiopatologia , Teste de Caminhada , Adulto Jovem
8.
Circulation ; 115(7): 896-908, 2007 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-17283259

RESUMO

BACKGROUND: Ex vivo expansion of resident cardiac stem cells, followed by delivery to the heart, may favor regeneration and functional improvement. METHODS AND RESULTS: Percutaneous endomyocardial biopsy specimens grown in primary culture developed multicellular clusters known as cardiospheres, which were plated to yield cardiosphere-derived cells (CDCs). CDCs from human biopsy specimens and from comparable porcine samples were examined in vitro for biophysical and cytochemical evidence of cardiogenic differentiation. In addition, human CDCs were injected into the border zone of acute myocardial infarcts in immunodeficient mice. Biopsy specimens from 69 of 70 patients yielded cardiosphere-forming cells. Cardiospheres and CDCs expressed antigenic characteristics of stem cells at each stage of processing, as well as proteins vital for cardiac contractile and electrical function. Human and porcine CDCs cocultured with neonatal rat ventricular myocytes exhibited biophysical signatures characteristic of myocytes, including calcium transients synchronous with those of neighboring myocytes. Human CDCs injected into the border zone of myocardial infarcts engrafted and migrated into the infarct zone. After 20 days, the percentage of viable myocardium within the infarct zone was greater in the CDC-treated group than in the fibroblast-treated control group; likewise, left ventricular ejection fraction was higher in the CDC-treated group. CONCLUSIONS: A method is presented for the isolation of adult human stem cells from endomyocardial biopsy specimens. CDCs are cardiogenic in vitro; they promote cardiac regeneration and improve heart function in a mouse infarct model, which provides motivation for further development for therapeutic applications in patients.


Assuntos
Células-Tronco Adultas/fisiologia , Infarto do Miocárdio/terapia , Miocárdio/patologia , Miócitos Cardíacos/fisiologia , Regeneração , Animais , Biópsia , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Humanos , Camundongos , Miocárdio/citologia , Ratos , Transplante de Células-Tronco , Suínos
9.
J Extracell Vesicles ; 6(1): 1347019, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28815002

RESUMO

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) is one of the most sensitive, economical and widely used methods for evaluating gene expression. However, the utility of this method continues to be undermined by a number of challenges including normalization using appropriate reference genes. The need to develop tailored and effective strategies is further underscored by the burgeoning field of extracellular vesicle (EV) biology. EVs contain unique signatures of small RNAs including microRNAs (miRs). In this study we develop and validate a comprehensive strategy for identifying highly stable reference genes in a therapeutically relevant cell type, cardiosphere-derived cells. Data were analysed using the four major approaches for reference gene evaluation: NormFinder, GeNorm, BestKeeper and the Delta Ct method. The weighted geometric mean of all of these methods was obtained for the final ranking. Analysis of RNA sequencing identified miR-101-3p, miR-23a-3p and a previously identified EV reference gene, miR-26a-5p. Analysis of a chip-based method (NanoString) identified miR-23a, miR-217 and miR-379 as stable candidates. RT-qPCR validation revealed that the mean of miR-23a-3p, miR-101-3p and miR-26a-5p was the most stable normalization strategy. Here, we demonstrate that a comprehensive approach of a diverse data set of conditions using multiple algorithms reliably identifies stable reference genes which will increase the utility of gene expression evaluation of therapeutically relevant EVs.

10.
JACC Heart Fail ; 2(1): 49-61, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24511463

RESUMO

OBJECTIVES: This study sought to compare the regenerative potency of cells derived from healthy and diseased human hearts. BACKGROUND: Results from pre-clinical studies and the CADUCEUS (CArdiosphere-Derived aUtologous stem CElls to reverse ventricUlar dySfunction) trial support the notion that cardiosphere-derived cells (CDCs) from normal and recently infarcted hearts are capable of regenerating healthy heart tissue after myocardial infarction (MI). It is unknown whether CDCs derived from advanced heart failure (HF) patients retain the same regenerative potency. METHODS: In a mouse model of acute MI, we compared the regenerative potential and functional benefits of CDCs derived from 3 groups: 1) non-failing (NF) donor: healthy donor hearts post-transplantation; 2) MI: patients who had an MI 9 to 35 days before biopsy; and 3) HF: advanced cardiomyopathy tissue explanted at cardiac transplantation. RESULTS: Cell growth and phenotype were identical in all 3 groups. Injection of HF CDCs led to the greatest therapeutic benefit in mice, with the highest left ventricular ejection fraction, thickest infarct wall, most viable tissue, and least scar 3 weeks after treatment. In vitro assays revealed that HF CDCs secreted higher levels of stromal cell-derived factor (SDF)-1, which may contribute to the cells' augmented resistance to oxidative stress, enhanced angiogenesis, and improved myocyte survival. Histological analysis indicated that HF CDCs engrafted better, recruited more endogenous stem cells, and induced greater angiogenesis and cardiomyocyte cell-cycle re-entry. CDC-secreted SDF-1 levels correlated with decreases in scar mass over time in CADUCEUS patients treated with autologous CDCs. CONCLUSIONS: CDCs from advanced HF patients exhibit augmented potency in ameliorating ventricular dysfunction post-MI, possibly through SDF-1­mediated mechanisms.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Coração/fisiologia , Miócitos Cardíacos/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Adulto , Idoso , Animais , Cardiomiopatias/fisiopatologia , Matriz Extracelular , Feminino , Sobrevivência de Enxerto/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos SCID , Pessoa de Meia-Idade , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica/fisiologia , Estresse Oxidativo/fisiologia , Transplante de Células-Tronco/métodos , Transplante Autólogo
11.
Biomatter ; 3(1)2013.
Artigo em Inglês | MEDLINE | ID: mdl-23538511

RESUMO

Cardiosphere-derived cells (CDCs) are under clinical development and are currently being tested in a clinical trial enrolling patients who have undergone a myocardial infarction. CDCs are presently administered via infusion into the infarct-related artery and have been shown in early clinical trials to be effective agents of myocardial regeneration. This review describes the administration of CDCs in a hyaluronan-gelatin hydrogel via myocardial injection and the subsequent improvements in therapeutic benefit seen in animal models. Development of a next generation therapy involving the combination of CDCs and hydrogel is discussed.


Assuntos
Gelatina/química , Ácido Hialurônico/administração & dosagem , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Infarto do Miocárdio/terapia , Miócitos Cardíacos/transplante , Animais , Terapia Baseada em Transplante de Células e Tecidos , Ensaios Clínicos como Assunto , Gelatina/administração & dosagem , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/administração & dosagem , Infarto do Miocárdio/patologia , Miócitos Cardíacos/citologia
12.
Expert Rev Cardiovasc Ther ; 10(9): 1185-94, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23098154

RESUMO

Heart disease is a major cause of morbidity and mortality. Cellular therapies hold significant promise for patients with heart disease. Heart-derived progenitor cells are capable of repairing a diseased heart through modulation of growth factor milieu and temporary engraftment leading to endogenous repair. The proof-of-concept CADUCEUS clinical trial using cardiosphere-derived cells has shown evidence of therapeutic cardiac tissue regeneration. Future clinical trials are now being planned to generate additional safety and efficacy data in the hopes of building toward an approved cellular therapy for heart disease.


Assuntos
Células-Tronco Adultas/efeitos dos fármacos , Células-Tronco Adultas/transplante , Albuminas/uso terapêutico , Infarto do Miocárdio/terapia , Poliésteres/uso terapêutico , Albuminas/administração & dosagem , Albuminas/efeitos adversos , Albuminas/farmacologia , Animais , Cateteres Cardíacos , Coração/efeitos dos fármacos , Coração/fisiologia , Humanos , Infusões Intra-Arteriais , Injeções Intramusculares , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Miocárdio/citologia , Poliésteres/administração & dosagem , Poliésteres/efeitos adversos , Poliésteres/farmacologia , Regeneração , Transplante de Células-Tronco/efeitos adversos , Transplante de Células-Tronco/métodos
13.
J Am Coll Cardiol ; 59(10): 942-53, 2012 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-22381431

RESUMO

OBJECTIVES: The goal of this study was to conduct a direct head-to-head comparison of different stem cell types in vitro for various assays of potency and in vivo for functional myocardial repair in the same mouse model of myocardial infarction. BACKGROUND: Adult stem cells of diverse origins (e.g., bone marrow, fat, heart) and antigenic identity have been studied for repair of the damaged heart, but the relative utility of the various cell types remains unclear. METHODS: Human cardiosphere-derived cells (CDCs), bone marrow-derived mesenchymal stem cells, adipose tissue-derived mesenchymal stem cells, and bone marrow mononuclear cells were compared. RESULTS: CDCs revealed a distinctive phenotype with uniform expression of CD105, partial expression of c-kit and CD90, and negligible expression of hematopoietic markers. In vitro, CDCs showed the greatest myogenic differentiation potency, highest angiogenic potential, and relatively high production of various angiogenic and antiapoptotic-secreted factors. In vivo, injection of CDCs into the infarcted mouse hearts resulted in superior improvement of cardiac function, the highest cell engraftment and myogenic differentiation rates, and the least-abnormal heart morphology 3 weeks after treatment. CDC-treated hearts also exhibited the lowest number of apoptotic cells. The c-kit(+) subpopulation purified from CDCs produced lower levels of paracrine factors and inferior functional benefit when compared with unsorted CDCs. To validate the comparison of cells from various human donors, selected results were confirmed in cells of different types derived from individual rats. CONCLUSIONS: CDCs exhibited a balanced profile of paracrine factor production and, among various comparator cell types/subpopulations, provided the greatest functional benefit in experimental myocardial infarction.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Mesenquimais/citologia , Infarto do Miocárdio/terapia , Miocárdio/citologia , Miócitos Cardíacos/transplante , Comunicação Parácrina/fisiologia , Transplante de Células-Tronco , Albuminas/farmacologia , Animais , Apoptose , Diferenciação Celular , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos SCID , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/citologia , Poliésteres/farmacologia , Ratos , Ratos Endogâmicos WKY , Cicatrização
14.
Heart Rhythm ; 5(6): 880-7, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18534373

RESUMO

New approaches for cardiac repair have been enabled by the discovery that the heart contains its own reservoir of stem cells. In Part 1 of this review, we discussed various cardiac stem cell populations, reviewed our own work on cardiosphere-derived cells from human hearts, and outlined large animal preclinical models testing the regenerative potential of cardiac stem cells. Here we continue with a discussion on other adult stem cell sources with clinical potential. We summarize the critical safety issues associated with stem cell therapy and present the possible proarrhythmic and antiarrhythmic effects of stem cell transplantation. We discuss the outcomes of clinical stem cell trials and identify the technical, ethical, and practical issues facing the clinical application of cardiac stem cells.


Assuntos
Arritmias Cardíacas/etiologia , Ensaios Clínicos como Assunto , Infarto do Miocárdio , Miocárdio , Transplante de Células-Tronco/efeitos adversos , Arritmias Cardíacas/fisiopatologia , Fusão Celular , Ensaios Clínicos como Assunto/tendências , Endotélio Vascular , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Músculo Esquelético/citologia , Mioblastos , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/citologia , Células-Tronco , Transplante Autólogo/efeitos adversos , Resultado do Tratamento
15.
Heart Rhythm ; 5(5): 749-57, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18452881

RESUMO

New approaches for cardiac repair have been enabled by the discovery that the heart contains its own reservoir of stem cells. These cells are positive for various stem/progenitor cell markers, are self-renewing, and exhibit multilineage differentiation potential. Recently we developed a method for ex vivo expansion of cardiac-derived stem cells from human myocardial biopsies with a view to subsequent autologous transplantation for myocardial regeneration. Here we review the state of the cardiac stem cell field and our own work on cardiosphere-derived stem cells from human hearts. The first of this two-part review outlines emerging preclinical data on the application of cardiac stem cells. Part 2 continues with a discussion of other stem cell sources with clinical potential, a summary of the critical issues surrounding stem cell therapy (with an emphasis on the crucial issue of how cell transplantation may influence arrhythmias), our perception of clinical stem cell trials to date, and the issues facing the clinical application of cardiac stem cells.


Assuntos
Cardiopatias/terapia , Células-Tronco/classificação , Humanos , Transplante de Células-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA