Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
2.
PLoS One ; 14(12): e0227283, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31891633

RESUMO

Upon myocardial damage, the release of cardiac proteins induces a strong antibody-mediated immune response, which can lead to adverse cardiac remodeling and eventually heart failure (HF). Stem cell therapy using mesenchymal stromal cells (MSCs) or cardiomyocyte progenitor cells (CPCs) previously showed beneficial effects on cardiac function despite low engraftment in the heart. Paracrine mediators are likely of great importance, where, for example, MSC-derived extracellular vesicles (EVs) also show immunosuppressive properties in vitro. However, the limited capacity of MSCs to differentiate into cardiac cells and the sufficient scaling of MSC-derived EVs remain a challenge to clinical translation. Therefore, we investigated the immunosuppressive actions of endogenous CPCs and CPC-derived EVs on antibody production in vitro, using both healthy controls and end-stage HF patients. Both MSCs and CPCs strongly inhibit lymphocyte proliferation and antibody production in vitro. Furthermore, CPC-derived EVs significantly lowered the levels of IgG1, IgG4, and IgM, especially when administered for longer duration. In line with previous findings, plasma cells of end-stage HF patients showed high production of IgG3, which can be inhibited by MSCs in vitro. MSCs and CPCs inhibit in vitro antibody production of both healthy and end-stage HF-derived immune cells. CPC-derived paracrine factors, such as EVs, show similar effects, but do not provide the complete immunosuppressive capacity of CPCs. The strongest immunosuppressive effects were observed using MSCs, suggesting that MSCs might be the best candidates for therapeutic targeting of B-cell responses in HF.


Assuntos
Linfócitos B/imunologia , Insuficiência Cardíaca/terapia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Transplante de Células-Tronco Mesenquimais , Mioblastos Cardíacos/transplante , Linfócitos B/citologia , Proliferação de Células , Células Cultivadas , Vesículas Extracelulares/imunologia , Insuficiência Cardíaca/imunologia , Humanos
3.
Anatol J Cardiol ; 20(6): 318-329, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30504732

RESUMO

OBJECTIVE: To evaluate the effects of transplantation of hypoxia-inducible factor-1α (HIF-1α) gene-modified cardiac stem cells (CSCs) on the cardiac function of heart failure rats after myocardial infarction (MI). METHODS: Twenty-four Sprague-Dawley rats were randomly divided into three groups: HIF-1α-modified CSCs group, single CSCs group, and model group. The model of heart failure after MI was established by thoracotomy-left anterior descending coronary artery ligation, followed by injection of modified CSCs, single CSCs, and PBS, respectively, 2 weeks later. The results were observed 4 weeks later. RESULTS: CSCs were infected with recombinant adenovirus. HIF-1α mRNA level of HIF-1α-enhanced green fluorescent protein (EGFP)+CSCs group significantly surpassed those of EGFP+CSCs and CSCs groups (p<0.001). Left ventricular ejection fractions (LVEFs) of HIF-1α+CSCs+MI and CSCs+MI groups significantly increased compared with the model group (p<0.001). The difference between LVEFs before and after transplantation was positively correlated with the survival rate of CSCs in infarction border zone (r=0.867, p<0.001). The apoptosis rate of HIF1α+CSCs+MI group was significantly lower than that of CSCs+MI group (p=0.008). The expression of vascular endothelial growth factor protein in HIF-1α+CSCs+MI group significantly increased, compared with that of MI group (p<0.001). The capillary density of HIF-1α+CSCs+MI group significantly exceeded that of CSCs+MI group (p<0.001). CONCLUSION: Transplantation of either HIF-1α-modified CSCs or single CSCs reduced cardiomyocyte apoptosis in rats with heart failure after MI, promoted vascular regeneration in infarct area, and improved cardiac function. Particularly, HIF-1α-modified CSCs had more significant effects.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Coração/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Mioblastos Cardíacos/transplante , Infarto do Miocárdio/complicações , Adenoviridae , Animais , Apoptose , Técnicas de Cultura de Células , Engenharia Genética , Coração/diagnóstico por imagem , Insuficiência Cardíaca/etiologia , Masculino , Neovascularização Fisiológica , Ratos , Ratos Sprague-Dawley , Transfecção , Ultrassonografia , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
JCI Insight ; 3(19)2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30282820

RESUMO

Sudden death is the most common mode of exodus in patients with heart failure and preserved ejection fraction (HFpEF). Cardiosphere-derived cells (CDCs) reduce inflammation and fibrosis in a rat model of HFpEF, improving diastolic function and prolonging survival. We tested the hypothesis that CDCs decrease ventricular arrhythmias (VAs) and thereby possibly contribute to prolonged survival. Dahl salt-sensitive rats were fed a high-salt diet to induce HFpEF. Allogeneic rat CDCs (or phosphate-buffered saline as placebo) were injected in rats with echo-verified HFpEF. CDC-injected HFpEF rats were less prone to VA induction by programmed electrical stimulation. Action potential duration (APD) was shortened, and APD homogeneity was increased by CDC injection. Transient outward potassium current density was upregulated in cardiomyocytes from CDC rats relative to placebo, as were the underlying transcript (Kcnd3) and protein (Kv4.3) levels. Fibrosis was attenuated in CDC-treated hearts, and survival was increased. Sudden death risk also trended down, albeit nonsignificantly. CDC therapy decreased VA in HFpEF rats by shortening APD, improving APD homogeneity, and decreasing fibrosis. Unlike other stem/progenitor cells, which often exacerbate arrhythmias, CDCs reverse electrical remodeling and suppress arrhythmogenesis in HFpEF.


Assuntos
Potenciais de Ação , Arritmias Cardíacas/prevenção & controle , Morte Súbita Cardíaca/prevenção & controle , Insuficiência Cardíaca/mortalidade , Mioblastos Cardíacos/transplante , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/mortalidade , Morte Súbita Cardíaca/etiologia , Modelos Animais de Doenças , Ecocardiografia , Eletrocardiografia , Insuficiência Cardíaca/etiologia , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/fisiopatologia , Humanos , Masculino , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Ratos , Ratos Endogâmicos Dahl , Canais de Potássio Shal/metabolismo , Sódio na Dieta/efeitos adversos , Volume Sistólico , Transplante Homólogo , Remodelação Ventricular
5.
Circ Res ; 123(5): 579-589, 2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-29921651

RESUMO

RATIONALE: Allogeneic cardiac stem cells (AlloCSC-01) have shown protective, immunoregulatory, and regenerative properties with a robust safety profile in large animal models of heart disease. OBJECTIVE: To investigate the safety and feasibility of early administration of AlloCSC-01 in patients with ST-segment-elevation myocardial infarction. METHODS AND RESULTS: CAREMI (Safety and Efficacy of Intracoronary Infusion of Allogeneic Human Cardiac Stem Cells in Patients With STEMI and Left Ventricular Dysfunction) was a phase I/II multicenter, randomized, double-blind, placebo-controlled trial in patients with ST-segment-elevation myocardial infarction, left ventricular ejection fraction ≤45%, and infarct size ≥25% of left ventricular mass by cardiac magnetic resonance, who were randomized (2:1) to receive AlloCSC-01 or placebo through the intracoronary route at days 5 to 7. The primary end point was safety and included all-cause death and major adverse cardiac events at 30 days (all-cause death, reinfarction, hospitalization because of heart failure, sustained ventricular tachycardia, ventricular fibrillation, and stroke). Secondary safety end points included major adverse cardiac events at 6 and 12 months, adverse events, and immunologic surveillance. Secondary exploratory efficacy end points were changes in infarct size (percentage of left ventricular mass) and indices of ventricular remodeling by magnetic resonance at 12 months. Forty-nine patients were included (92% male, 55±11 years), 33 randomized to AlloCSC-01 and 16 to placebo. No deaths or major adverse cardiac events were reported at 12 months. One severe adverse events in each group was considered possibly related to study treatment (allergic dermatitis and rash). AlloCSC-01 elicited low levels of donor-specific antibodies in 2 patients. No immune-related adverse events were found, and no differences between groups were observed in magnetic resonance-based efficacy parameters at 12 months. The estimated treatment effect of AlloCSC-01 on the absolute change from baseline in infarct size was -2.3% (95% confidence interval, -6.5% to 1.9%). CONCLUSIONS: AlloCSC-01 can be safely administered in ST-segment-elevation myocardial infarction patients with left ventricular dysfunction early after revascularization. Low immunogenicity and absence of immune-mediated events will facilitate adequately powered studies to demonstrate their clinical efficacy in this setting. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov . Unique identifier: NCT02439398.


Assuntos
Mioblastos Cardíacos/transplante , Infarto do Miocárdio/terapia , Transplante de Células-Tronco/métodos , Disfunção Ventricular Esquerda/terapia , Idoso , Feminino , Humanos , Infusões Intra-Arteriais , Masculino , Pessoa de Meia-Idade , Mioblastos Cardíacos/citologia , Infarto do Miocárdio/complicações , Transplante de Células-Tronco/efeitos adversos , Transplante Homólogo , Disfunção Ventricular Esquerda/complicações
6.
Circ Res ; 122(7): 958-969, 2018 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-29343525

RESUMO

RATIONALE: Human pluripotent stem cell-derived cardiovascular progenitor cells (hPSC-CVPCs) should be thoroughly investigated in large animal studies before testing in clinical trials. OBJECTIVE: The main of this study is to clarify whether hPSC-CVPCs can engraft for long time in the heart of primates after myocardial infarction (MI) and compare the effectiveness and safety of immunosuppression with cyclosporine alone or multiple-drug regimen (MDR) containing cyclosporine, methylprednisolone, and basiliximab in cynomolgus monkeys that had received intramyocardial injections of 1×107 EGFP (enhanced green fluorescent protein)-expressing hPSC-CVPCs after MI. A third group of animals received the immunosuppression MDR but without cell therapy after MI (MI+MDR group). METHODS AND RESULTS: Measurements of EGFP gene levels and EGFP immunofluorescence staining indicated that the hPSC-CVPC engraftment rate was greater in the MI+MDR+CVPC group than that in the MI+cyclosporine+CVPC group. However, even in the MI+MDR+CVPC group, no transplanted cells could be detected at 140 days after transplantation. Concomitantly, immunofluorescent analysis of CD3, CD4, and CD8 expression indicated that T-lymphocyte infiltration in the CVPC-transplanted hearts was less in the MDR-treated animals than in the cyclosporine-alone-treated animals. The recovery of left ventricular function on day 28 post-MI in the MI+MDR+CVPC group was better than that in the MI+MDR group. Apoptotic cardiac cells were also less common in the MI+MDR+CVPC group than in the MI+MDR group, although both immunosuppression regimens were associated with transient hepatic dysfunction. CONCLUSIONS: This is the largest study of hPSCs in nonhuman primates in cardiovascular field to date (n=32). Compared with cyclosporine alone, MDR attenuates immune rejection and improves survival of hPSC-CVPCs in primates; this is associated with less apoptosis of native cardiac cells and better recovery of left ventricular function at 28 days. However, even with MDR, transplanted hPSC-CVPCs do not engraft and do not survive at 140 days after transplantation, thereby excluding remuscularization as a mechanism for the functional effect.


Assuntos
Células-Tronco Embrionárias Humanas/citologia , Desenvolvimento Muscular , Mioblastos Cardíacos/transplante , Infarto do Miocárdio/terapia , Transplante de Células-Tronco/métodos , Animais , Linhagem Celular , Ciclosporina/administração & dosagem , Ciclosporina/efeitos adversos , Humanos , Terapia de Imunossupressão/efeitos adversos , Terapia de Imunossupressão/métodos , Imunossupressores/administração & dosagem , Imunossupressores/efeitos adversos , Macaca fascicularis , Masculino , Mioblastos Cardíacos/citologia , Transplante de Células-Tronco/efeitos adversos
7.
Circ Res ; 122(7): 994-1005, 2018 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-29367212

RESUMO

RATIONALE: Intracoronary administration of cardiosphere-derived cells (CDCs) in patients with single ventricles resulted in a short-term improvement in cardiac function. OBJECTIVE: To test the hypothesis that CDC infusion is associated with improved cardiac function and reduced mortality in patients with heart failure. METHODS AND RESULTS: We evaluated the effectiveness of CDCs using an integrated cohort study in 101 patients with single ventricles, including 41 patients who received CDC infusion and 60 controls treated with staged palliation alone. Heart failure with preserved ejection fraction (EF) or reduced EF was stratified by the cardiac function after surgical reconstruction. The main outcome measure was to evaluate the magnitude of improvement in cardiac function and all-cause mortality at 2 years. Animal studies were conducted to clarify the underlying mechanisms of heart failure with preserved EF and heart failure with reduced EF phenotypes. At 2 years, CDC infusion increased ventricular function (stage 2: +8.4±10.0% versus +1.6±6.4%, P=0.03; stage 3: +7.9±7.5% versus -1.1±5.5%, P<0.001) compared with controls. In all available follow-up data, survival did not differ between the 2 groups (log-rank P=0.225), whereas overall patients treated by CDCs had lower incidences of late failure (P=0.022), adverse events (P=0.013), and catheter intervention (P=0.005) compared with controls. CDC infusion was associated with a lower risk of adverse events (hazard ratio, 0.411; 95% CI, 0.179-0.942; P=0.036). Notably, CDC infusion reduced mortality (P=0.038) and late complications (P<0.05) in patients with heart failure with reduced EF but not with heart failure with preserved EF. CDC-treated rats significantly reversed myocardial fibrosis with differential collagen deposition and inflammatory responses between the heart failure phenotypes. CONCLUSIONS: CDC administration in patients with single ventricles showed favorable effects on ventricular function and was associated with reduced late complications except for all-cause mortality after staged procedures. Patients with heart failure with reduced EF but not heart failure with preserved EF treated by CDCs resulted in significant improvement in clinical outcome. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifiers: NCT01273857 and NCT01829750.


Assuntos
Cardiopatias Congênitas/terapia , Insuficiência Cardíaca/terapia , Ventrículos do Coração/anormalidades , Mioblastos Cardíacos/transplante , Transplante de Células-Tronco/métodos , Pré-Escolar , Feminino , Cardiopatias Congênitas/complicações , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/prevenção & controle , Hemodinâmica , Humanos , Lactente , Masculino , Transplante de Células-Tronco/efeitos adversos , Função Ventricular
8.
Stem Cell Res Ther ; 8(1): 230, 2017 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-29037258

RESUMO

BACKGROUND: Application of cardiac stem cells combined with biomaterial scaffold is a promising therapeutic strategy for heart repair after myocardial infarction. However, the optimal cell types and biomaterials remain elusive. METHODS: In this study, we seeded Isl1+ embryonic cardiac progenitor cells (CPCs) into decellularized porcine small intestinal submucosa extracellular matrix (SIS-ECM) to assess the therapeutic potential of Isl1+ CPCs and the biocompatibility of SIS-ECM with these cells. RESULTS: We observed that SIS-ECM supported the viability and attachment of Isl1+ CPCs. Importantly, Isl1+ CPCs differentiated into cardiomyocytes and endothelial cells 7 days after seeding into SIS-ECM. In addition, SIS-ECM with CPC-derived cardiomyocytes showed spontaneous contraction and responded to ß-adrenergic stimulation. Next, patches of SIS-ECM seeded with CPCs for 7 days were transplanted onto the outer surface of infarcted myocardium in mice. Four weeks after transplantation, the patches were tightly attached to the surface of the host myocardium and remained viable. Transplantation of patches improved cardiac function, decreased the left ventricular myocardial scarring area, and reduced fibrosis and heart failure. CONCLUSIONS: Transplantation of Isl1+ CPCs seeded in SIS-ECM represents an effective approach for cell-based heart therapy.


Assuntos
Mucosa Intestinal/metabolismo , Mioblastos Cardíacos/transplante , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular , Células Cultivadas , Matriz Extracelular/metabolismo , Intestino Delgado/metabolismo , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/metabolismo , Miócitos Cardíacos/metabolismo , Suínos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Adv Drug Deliv Rev ; 106(Pt A): 104-115, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27133386

RESUMO

Stem cell-based therapies form an exciting new class of medicine that attempt to provide the body with the building blocks required for the reconstruction of damaged organs. However, delivering cells to the correct location, while preserving their integrity and functional properties, is a complex undertaking. These challenges have led to the development of a highly dynamic interdisciplinary research field, wherein medical, biological, and chemical sciences have collaborated to develop strategies to overcome the physiological barriers imposed on the cellular therapeutics. In this respect, improving the acute retention and subsequent survival of stem cells is key to effectively increase the effect of the therapy, while proper tissue integration is imperative for stem cells to functionally replace lost cells in damaged organs. In this review, we will use the heart as an example to highlight the current knowledge of therapeutic stem cell utilization, the existing pitfalls and limitations, and the approaches that have been developed to overcome them.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/transplante , Transplante de Células-Tronco/métodos , Animais , Humanos
12.
Cell Stem Cell ; 18(3): 368-81, 2016 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-26942852

RESUMO

Stem cell-based approaches to cardiac regeneration are increasingly viable strategies for treating heart failure. Generating abundant and functional autologous cells for transplantation in such a setting, however, remains a significant challenge. Here, we isolated a cell population with extensive proliferation capacity and restricted cardiovascular differentiation potentials during cardiac transdifferentiation of mouse fibroblasts. These induced expandable cardiovascular progenitor cells (ieCPCs) proliferated extensively for more than 18 passages in chemically defined conditions, with 10(5) starting fibroblasts robustly producing 10(16) ieCPCs. ieCPCs expressed cardiac signature genes and readily differentiated into functional cardiomyocytes (CMs), endothelial cells (ECs), and smooth muscle cells (SMCs) in vitro, even after long-term expansion. When transplanted into mouse hearts following myocardial infarction, ieCPCs spontaneously differentiated into CMs, ECs, and SMCs and improved cardiac function for up to 12 weeks after transplantation. Thus, ieCPCs are a powerful system to study cardiovascular specification and provide strategies for regenerative medicine in the heart.


Assuntos
Técnicas de Reprogramação Celular , Reprogramação Celular , Fibroblastos , Células-Tronco Pluripotentes Induzidas , Mioblastos Cardíacos , Infarto do Miocárdio , Transplante de Células-Tronco , Animais , Fibroblastos/metabolismo , Fibroblastos/transplante , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/transplante , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/terapia
13.
Adv Healthc Mater ; 5(9): 1071-9, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26913710

RESUMO

Inadequate cell retention and survival in cardiac stem cell therapy seems to be reducing the therapeutic effect of the injected stem cells. In order to ameliorate their regenerative effects, various biomaterials are being investigated for their potential supportive properties. Here, gelatin microspheres (MS) are utilized as microcarriers to improve the delivery and therapeutic efficacy of cardiac progenitor cells (CPCs) in the ischemic myocardium. The gelatin MS, generated from a water-in-oil emulsion, are able to accommodate the attachment of CPCs, thereby maintaining their cardiogenic potential. In a mouse model of myocardial infarction, we demonstrated the ability of these microcarriers to substantially enhance cell engraftment in the myocardium as indicated by bioluminescent imaging and histological analysis. However, despite an observed tenfold increase in CPC numbers in the myocardium, echocardiography, and histology reveals that mice treated with MS-CPCs show marginal improvement in cardiac function compared to CPCs only. Overall, a straightforward and translational approach is developed to increase the retention of stem cells in the ischemic myocardium. Even though the current biomaterial setup with CPCs as cell source does not translate into improved therapeutic action, coupling this developed technology with stem cell-derived cardiomyocytes can lead to an effective remuscularization therapy.


Assuntos
Células Imobilizadas , Gelatina/química , Microesferas , Mioblastos Cardíacos , Infarto do Miocárdio/terapia , Miocárdio/metabolismo , Animais , Células Imobilizadas/metabolismo , Células Imobilizadas/transplante , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/transplante , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia
14.
Stem Cells Transl Med ; 5(1): 67-74, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26582908

RESUMO

UNLABELLED: Given the limited regenerative capacity of the heart, cellular therapy with stem cell-derived cardiac cells could be a potential treatment for patients with heart disease. However, reliable imaging techniques to longitudinally assess engraftment of the transplanted cells are scant. To address this issue, we used ferumoxytol as a labeling agent of human embryonic stem cell-derived cardiac progenitor cells (hESC-CPCs) to facilitate tracking by magnetic resonance imaging (MRI) in a large animal model. Differentiating hESCs were exposed to ferumoxytol at different time points and varying concentrations. We determined that treatment with ferumoxytol at 300 µg/ml on day 0 of cardiac differentiation offered adequate cell viability and signal intensity for MRI detection without compromising further differentiation into definitive cardiac lineages. Labeled hESC-CPCs were transplanted by open surgical methods into the left ventricular free wall of uninjured pig hearts and imaged both ex vivo and in vivo. Comprehensive T2*-weighted images were obtained immediately after transplantation and 40 days later before termination. The localization and dispersion of labeled cells could be effectively imaged and tracked at days 0 and 40 by MRI. Thus, under the described conditions, ferumoxytol can be used as a long-term, differentiation-neutral cell-labeling agent to track transplanted hESC-CPCs in vivo using MRI. SIGNIFICANCE: The development of a safe and reproducible in vivo imaging technique to track the fate of transplanted human embryonic stem cell-derived cardiac progenitor cells (hESC-CPCs) is a necessary step to clinical translation. An iron oxide nanoparticle (ferumoxytol)-based approach was used for cell labeling and subsequent in vivo magnetic resonance imaging monitoring of hESC-CPCs transplanted into uninjured pig hearts. The present results demonstrate the use of ferumoxytol labeling and imaging techniques in tracking the location and dispersion of cell grafts, highlighting its utility in future cardiac stem cell therapy trials.


Assuntos
Rastreamento de Células/métodos , Células-Tronco Embrionárias , Óxido Ferroso-Férrico/farmacologia , Imageamento por Ressonância Magnética , Mioblastos Cardíacos , Transplante de Células-Tronco , Células-Tronco Embrionárias/diagnóstico por imagem , Células-Tronco Embrionárias/transplante , Compostos Férricos/farmacologia , Óxido Ferroso-Férrico/farmacocinética , Xenoenxertos , Humanos , Mioblastos Cardíacos/diagnóstico por imagem , Mioblastos Cardíacos/transplante , Radiografia
16.
J Am Coll Cardiol ; 66(18): 1990-1999, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26516002

RESUMO

BACKGROUND: Both bone marrow-derived mesenchymal stem cells (MSCs) and c-kit(+) cardiac stem cells (CSCs) improve left ventricular remodeling in porcine models and clinical trials. Using xenogeneic (human) cells in immunosuppressed animals with acute ischemic heart disease, we previously showed that these 2 cell types act synergistically. OBJECTIVES: To more accurately model clinical applications for heart failure, this study tested whether the combination of autologous MSCs and CSCs produce greater improvement in cardiac performance than MSCs alone in a nonimmunosuppressed porcine model of chronic ischemic cardiomyopathy. METHODS: Three months after ischemia/reperfusion injury, Göttingen swine received transendocardial injections with MSCs alone (n = 6) or in combination with cardiac-derived CSCs (n = 8), or placebo (vehicle; n = 6). Cardiac functional and anatomic parameters were assessed using cardiac magnetic resonance at baseline and before and after therapy. RESULTS: Both groups of cell-treated animals exhibited significantly reduced scar size (MSCs -44.1 ± 6.8%; CSC/MSC -37.2 ± 5.4%; placebo -12.9 ± 4.2%; p < 0.0001), increased viable tissue, and improved wall motion relative to placebo 3 months post-injection. Ejection fraction (EF) improved (MSCs 2.9 ± 1.6 EF units; CSC/MSC 6.9 ± 2.8 EF units; placebo 2.5 ± 1.6 EF units; p = 0.0009), as did stroke volume, cardiac output, and diastolic strain only in the combination-treated animals, which also exhibited increased cardiomyocyte mitotic activity. CONCLUSIONS: These findings illustrate that interactions between MSCs and CSCs enhance cardiac performance more than MSCs alone, establish the safety of autologous cell combination strategies, and support the development of second-generation cell therapeutic products.


Assuntos
Cardiomiopatias , Transplante de Células-Tronco Mesenquimais/métodos , Mioblastos Cardíacos/transplante , Traumatismo por Reperfusão Miocárdica/complicações , Animais , Cardiomiopatias/diagnóstico , Cardiomiopatias/etiologia , Cardiomiopatias/fisiopatologia , Cardiomiopatias/terapia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Humanos , Imagem Cinética por Ressonância Magnética/métodos , Volume Sistólico , Suínos , Transplante Heterotópico/métodos , Resultado do Tratamento , Remodelação Ventricular
17.
Philos Trans R Soc Lond B Biol Sci ; 370(1680): 20140373, 2015 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-26416682

RESUMO

Stem cell-based therapy is currently tested in several trials of chronic heart failure. The main question is to determine how its implementation could be extended to common clinical practice. To fill this gap, it is critical to first validate the hypothesis that the grafted stem cells primarily act by harnessing endogenous repair pathways. The confirmation of this mechanism would have three major clinically relevant consequences: (i) the use of cardiac-committed cells, since even though cells primarily act in a paracrine manner, such a phenotype seems the most functionally effective; (ii) the optimization of early cell retention, rather than of sustained cell survival, so that the cells reside in the target tissue long enough to deliver the factors underpinning their action; and (iii) the reliance on allogeneic cells, the expected rejection of which should only have to be delayed since a permanent engraftment would no longer be the objective. One step further, the long-term objective of cell therapy could be to use the cells exclusively for producing factors and then to only administer them to the patient. The production process would then be closer to that of a biological pharmaceutic, thereby facilitating an extended clinical use.


Assuntos
Insuficiência Cardíaca/terapia , Transplante de Células-Tronco , Técnicas de Cultura de Células , Linhagem da Célula , Sobrevivência Celular , Humanos , Modelos Cardiovasculares , Mioblastos Cardíacos/fisiologia , Mioblastos Cardíacos/transplante , Transplante Autólogo , Transplante Homólogo
18.
Circ J ; 79(7): 1422-30, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26073608

RESUMO

Over the past 2 decades, cardiac regeneration has evolved from an exotic fringe of cardiovascular biology to the forefront of molecular, genetic, epigenetic, translational, and clinical investigations. The unmet patient need is the paucity of self-repair following infarction. Robust regeneration seen in models such as zebrafish and newborn mice has inspired the field, along with encouragement from modern methods that make even low levels of restorative growth discernible, changing the scientific and technical landscape for effective counter-measures. Approaches under study to augment cardiac repair complement each other, and encompass grafting cells of diverse kinds, restarting the cell cycle in post-mitotic ventricular myocytes, reprogramming non-myocytes, and exploiting the dormant progenitor/stem cells that lurk within the adult heart. The latter are the emphasis of the present review. Cardiac-resident stem cells (CSC) can be harvested from heart tissue, expanded, and delivered to the myocardium as a therapeutic product, whose benefits may be hoped to surpass those achieved in human trials of bone marrow. However, important questions are prompted by such cells' discovery. How do they benefit recipient hearts? Do they contribute, measurably, as an endogenous population, to self-repair? Even if "no," might CSCs be targets for activation in situ by growth factors and other developmental catalysts? And, what combination of distinguishing markers best demarcates the cells with robust clonal growth and cardiogenic potential?


Assuntos
Células-Tronco Adultas/citologia , Mioblastos Cardíacos/citologia , Adulto , Células-Tronco Adultas/fisiologia , Células-Tronco Adultas/transplante , Animais , Biomarcadores , Diferenciação Celular , Linhagem da Célula , Terapia Baseada em Transplante de Células e Tecidos , Perfilação da Expressão Gênica , Coração/fisiologia , Humanos , Camundongos , Modelos Animais , Modelos Cardiovasculares , Mioblastos Cardíacos/fisiologia , Mioblastos Cardíacos/transplante , Regeneração/fisiologia , Peixe-Zebra
19.
Bull Exp Biol Med ; 157(1): 143-5, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24909728

RESUMO

The use of triple systemic transplantation of cardiomyoblasts raised from the culture of allogenic bone marrow mesenchymal stem cells of a healthy donor according to the new medical technology licensed by Federal Service on Surveillance in Healthcare in the therapy of a patient with late radiation cardiomyopathy and radiation exudative pericarditis developed 45 years after radiation therapy for Hodgkin lymphoma. High efficiency of systemic transplantation of mesenchymal stem cells partially differentiated towards cardiomyocytes was demonstrated. The therapeutic effect persists for more than 2 years. Possible mechanisms of the therapeutic effect of this type of stem cells and the prospects of using cell therapy in the treatment of late radiation injuries of vital organs and tissues are discussed.


Assuntos
Cardiomiopatias/terapia , Transplante de Células-Tronco Mesenquimais , Mioblastos Cardíacos/transplante , Pericardite/terapia , Cardiomiopatias/diagnóstico por imagem , Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Diferenciação Celular , Raios gama/efeitos adversos , Doença de Hodgkin/complicações , Doença de Hodgkin/diagnóstico por imagem , Doença de Hodgkin/patologia , Doença de Hodgkin/radioterapia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Pessoa de Meia-Idade , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/fisiologia , Pericardite/diagnóstico por imagem , Pericardite/etiologia , Pericardite/patologia , Transplante Homólogo , Resultado do Tratamento , Ultrassonografia
20.
Cell Stem Cell ; 12(6): 689-98, 2013 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-23746978

RESUMO

Stem cell therapy for cardiac disease is an exciting but highly controversial research area. Strategies such as cell transplantation and reprogramming have demonstrated both intriguing and sobering results. Yet as clinical trials proceed, our incomplete understanding of stem cell behavior is made evident by numerous unresolved matters, such as the mechanisms of cardiomyocyte turnover or the optimal therapeutic strategies to achieve clinical efficacy. In this Perspective, we consider how cardiac stem cell biology has led us into clinical trials, and we suggest that achieving true cardiac regeneration in patients may ultimately require resolution of critical controversies in experimental cardiac regeneration.


Assuntos
Cardiopatias/terapia , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/transplante , Miocárdio/citologia , Regeneração , Cardiopatias/patologia , Humanos , Miocárdio/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...