Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res Ther ; 7(1): 60, 2016 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-27225482

RESUMO

BACKGROUND: Although patient-sourced cardiac stem cells repair damaged myocardium, the extent to which medical co-morbidities influence cardiac-derived cell products is uncertain. Therefore, we investigated the influence of atherosclerotic risk factors on the regenerative performance of human cardiac explant-derived cells (EDCs). METHODS: In this study, the Long Term Stratification for survivors of acute coronary syndromes model was used to quantify the burden of cardiovascular risk factors within a group of patients with established atherosclerosis. EDCs were cultured from human atrial appendages and injected into immunodeficient mice 7 days post-left coronary ligation. Cytokine arrays and enzyme linked immunoassays were used to determine the release of cytokines by EDCs in vitro, and echocardiography was used to determine regenerative capabilities in vivo. RESULTS: EDCs sourced from patients with more cardiovascular risk factors demonstrated a negative correlation with production of pro-healing cytokines (such as stromal cell derived factor 1α) and exosomes which had negative effects on the promotion of angiogenesis and chemotaxis. Reductions in exosomes and pro-healing cytokines with accumulating medical co-morbidities were associated with increases in production of the pro-inflammatory cytokine interleukin-6 (IL-6) by EDCs. Increased patient co-morbidities were also correlated with significant attenuation in improvements of left ventricular ejection fraction. CONCLUSIONS: The regenerative performance of the earliest precursor cell population cultured from human explant tissue declines with accumulating medical co-morbidities. This effect is associated with diminished production of pro-cardiogenic cytokines and exosomes while IL-6 is markedly increased. Predictors of cardiac events demonstrated a lower capacity to support angiogenesis and repair injured myocardium in a mouse model of myocardial infarction.


Assuntos
Síndrome Coronariana Aguda/patologia , Aterosclerose/patologia , Estenose Coronária/patologia , Infarto do Miocárdio/patologia , Transplante de Células-Tronco , Síndrome Coronariana Aguda/metabolismo , Animais , Aterosclerose/metabolismo , Quimiocina CXCL12/biossíntese , Quimiocina CXCL12/metabolismo , Quimiotaxia , Comorbidade , Estenose Coronária/diagnóstico por imagem , Estenose Coronária/metabolismo , Modelos Animais de Doenças , Ecocardiografia , Exossomos/transplante , Expressão Gênica , Átrios do Coração/metabolismo , Átrios do Coração/patologia , Humanos , Interleucina-6/biossíntese , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/metabolismo , Neovascularização Fisiológica , Cultura Primária de Células , Células-Tronco/metabolismo , Células-Tronco/patologia , Volume Sistólico , Função Ventricular Esquerda
2.
Stem Cells ; 34(7): 1826-35, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27059540

RESUMO

First generation cardiac stem cell products provide indirect cardiac repair but variably produce key cardioprotective cytokines, such as stromal-cell derived factor 1α, which opens the prospect of maximizing up-front paracrine-mediated repair. The mesenchymal subpopulation within explant derived human cardiac stem cells underwent lentiviral mediated gene transfer of stromal-cell derived factor 1α. Unlike previous unsuccessful attempts to increase efficacy by boosting the paracrine signature of cardiac stem cells, cytokine profiling revealed that stromal-cell derived factor 1α over-expression prevented lv-mediated "loss of cytokines" through autocrine stimulation of CXCR4+ cardiac stem cells. Stromal-cell derived factor 1α enhanced angiogenesis and stem cell recruitment while priming cardiac stem cells to readily adopt a cardiac identity. As compared to injection with unmodified cardiac stem cells, transplant of stromal-cell derived factor 1α enhanced cells into immunodeficient mice improved myocardial function and angiogenesis while reducing scarring. Increases in myocardial stromal-cell derived factor 1α content paralleled reductions in myocyte apoptosis but did not influence long-term engraftment or the fate of transplanted cells. Transplantation of stromal-cell derived factor 1α transduced cardiac stem cells increased the generation of new myocytes, recruitment of bone marrow cells, new myocyte/vessel formation and the salvage of reversibly damaged myocardium to enhance cardiac repair after experimental infarction. Stem Cells 2016;34:1826-1835.


Assuntos
Quimiocina CXCL12/metabolismo , Miocárdio/citologia , Comunicação Parácrina , Células-Tronco/citologia , Células-Tronco/metabolismo , Engenharia Tecidual/métodos , Cicatrização , Animais , Diferenciação Celular , Humanos , Lentivirus/metabolismo , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Neovascularização Fisiológica , Receptores CXCR4 , Transdução Genética
3.
J Am Heart Assoc ; 4(9): e002104, 2015 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-26363004

RESUMO

BACKGROUND: Insulin-like growth factor 1 (IGF-1) activates prosurvival pathways and improves postischemic cardiac function, but this key cytokine is not robustly expressed by cultured human cardiac stem cells. We explored the influence of an enhanced IGF-1 paracrine signature on explant-derived cardiac stem cell-mediated cardiac repair. METHODS AND RESULTS: Receptor profiling demonstrated that IGF-1 receptor expression was increased in the infarct border zones of experimentally infarcted mice by 1 week after myocardial infarction. Human explant-derived cells underwent somatic gene transfer to overexpress human IGF-1 or the green fluorescent protein reporter alone. After culture in hypoxic reduced-serum media, overexpression of IGF-1 enhanced proliferation and expression of prosurvival transcripts and prosurvival proteins and decreased expression of apoptotic markers in both explant-derived cells and cocultured neonatal rat ventricular cardiomyocytes. Transplant of explant-derived cells genetically engineered to overexpress IGF-1 into immunodeficient mice 1 week after infarction boosted IGF-1 content within infarcted tissue and long-term engraftment of transplanted cells while reducing apoptosis and long-term myocardial scarring. CONCLUSIONS: Paracrine engineering of explant-derived cells to overexpress IGF-1 provided a targeted means of improving cardiac stem cell-mediated repair by enhancing the long-term survival of transplanted cells and surrounding myocardium.


Assuntos
Terapia Genética/métodos , Fator de Crescimento Insulin-Like I/biossíntese , Infarto do Miocárdio/cirurgia , Miocárdio/metabolismo , Comunicação Parácrina , Regeneração , Transplante de Células-Tronco , Células-Tronco/metabolismo , Idoso , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Humanos , Fator de Crescimento Insulin-Like I/genética , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Ratos , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Fatores de Tempo , Transfecção , Regulação para Cima
4.
Circulation ; 130(11 Suppl 1): S70-6, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25200058

RESUMO

BACKGROUND: The impact of diabetes mellitus on the cardiac regenerative potential of cardiac stem cells (CSCs) is unknown yet critical, given that individuals with diabetes mellitus may well require CSC therapy in the future. Using human and murine CSCs from diabetic cardiac tissue, we tested the hypothesis that hyperglycemic conditions impair CSC function. METHODS AND RESULTS: CSCs cultured from the cardiac biopsies of patients with diabetes mellitus (hemoglobin A1c, 10±2%) demonstrated reduced overall cell numbers compared with nondiabetic sourced biopsies (P=0.04). When injected into the infarct border zone of immunodeficient mice 1 week after myocardial infarction, CSCs from patients with diabetes mellitus demonstrated reduced cardiac repair compared with nondiabetic patients. Conditioned medium from CSCs of patients with diabetes mellitus displayed a reduced ability to promote in vitro blood vessel formation (P=0.02). Similarly, conditioned medium from CSCs cultured from the cardiac biopsies of streptozotocin-induced diabetic mice displayed impaired angiogenic capacity (P=0.0008). Somatic gene transfer of the methylglyoxal detoxification enzyme, glyoxalase-1, restored the angiogenic capacity of diabetic CSCs (diabetic transgenic versus nondiabetic transgenic; P=0.8). Culture of nondiabetic murine cardiac biopsies under high (25 mmol/L) glucose conditions reduced CSC yield (P=0.003), impaired angiogenic (P=0.02) and chemotactic (P=0.003) response, and reduced CSC-mediated cardiac repair (P<0.05). CONCLUSIONS: Diabetes mellitus reduces the ability of CSCs to repair injured myocardium. Both diabetes mellitus and preconditioning CSCs in high glucose attenuated the proangiogenic capacity of CSCs. Increased expression of glyoxalase-1 restored the proangiogenic capacity of diabetic CSCs, suggesting a means of reversing diabetic CSC dysfunction by interfering with the accumulation of reactive dicarbonyls.


Assuntos
Células-Tronco Adultas/transplante , Hiperglicemia/fisiopatologia , Células-Tronco Multipotentes/transplante , Neovascularização Fisiológica , Células-Tronco Adultas/efeitos dos fármacos , Animais , Apoptose , Biópsia , Células Cultivadas , Meios de Cultivo Condicionados , Diabetes Mellitus/patologia , Diabetes Mellitus Experimental/patologia , Genes Reporter , Humanos , Lactoilglutationa Liase/genética , Lactoilglutationa Liase/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Células-Tronco Multipotentes/efeitos dos fármacos , Miocárdio/patologia , Espécies Reativas de Oxigênio , Proteínas Recombinantes de Fusão/metabolismo
5.
Can J Cardiol ; 30(11): 1288-98, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25092406

RESUMO

Despite advances in treatment, heart failure remains one of the top killers in Canada. This recognition motivated a new research focus to harness the fundamental repair properties of the human heart. Since then, cardiac stem cells (CSCs) have emerged as a promising cell candidate to regenerate damaged hearts. The rationale of this approach is simple with ex vivo amplification of CSCs from clinical-grade biopsies, followed by delivery to areas of injury, where they engraft and regenerate the heart. In this review we will summarize recent advances and discuss future developments in CSC-mediated cardiac repair to treat the growing number of Canadians living with and dying from heart failure.


Assuntos
Insuficiência Cardíaca/terapia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Transplante de Células-Tronco/métodos , Diferenciação Celular , Células Cultivadas , Insuficiência Cardíaca/patologia , Humanos , Infarto do Miocárdio/patologia , Miocárdio
6.
Biomaterials ; 35(1): 133-42, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24099706

RESUMO

Transplantation of ex vivo proliferated cardiac stem cells (CSCs) is an emerging therapy for ischemic cardiomyopathy but outcomes are limited by modest engraftment and poor long-term survival. As such, we explored the effect of single cell microencapsulation to increase CSC engraftment and survival after myocardial injection. Transcript and protein profiling of human atrial appendage sourced CSCs revealed strong expression the pro-survival integrin dimers αVß3 and α5ß1- thus rationalizing the integration of fibronectin and fibrinogen into a supportive intra-capsular matrix. Encapsulation maintained CSC viability under hypoxic stress conditions and, when compared to standard suspended CSC, media conditioned by encapsulated CSCs demonstrated superior production of pro-angiogenic/cardioprotective cytokines, angiogenesis and recruitment of circulating angiogenic cells. Intra-myocardial injection of encapsulated CSCs after experimental myocardial infarction favorably affected long-term retention of CSCs, cardiac structure and function. Single cell encapsulation prevents detachment induced cell death while boosting the mechanical retention of CSCs to enhance repair of damaged myocardium.


Assuntos
Sobrevivência Celular , Coração/fisiopatologia , Hidrogéis , Isquemia Miocárdica/patologia , Miócitos Cardíacos/citologia , Células-Tronco/citologia , Idoso , Moléculas de Adesão Celular/metabolismo , Meios de Cultivo Condicionados , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/fisiopatologia , Miócitos Cardíacos/metabolismo , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...