Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cells Dev ; 28(9): 565-578, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30755138

RESUMO

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) hold great promise for regenerative medicine and in vitro screening. Despite displaying key cardiomyocyte phenotypic characteristics, they more closely resemble fetal/neonatal cardiomyocytes, and further characterization is necessary. By combining the use of tandem mass tags to label cell lysates, followed by multiplexing, we have determined the effects of short-term (30 day) in vitro culture on hiPSC-CM protein expression. We found that hiPSC-CM exhibit temporal changes in global protein expression; alterations in protein expression were pronounced during the first 2 weeks following thaw and dominated by reductions in proteins associated with protein synthesis and ubiquitination. Between 2 and 4 weeks, proceeding thaw alterations in protein expression were dominated by metabolic pathways, indicating a potential temporal metabolic shift from glycolysis toward oxidative phosphorylation. Time-dependent changes in proteins associated with cardiomyocyte contraction, excitation-contraction coupling, and metabolism were detected. While some were associated with expected functional outcomes in terms of morphology or electrophysiology, others such as metabolism did not produce the anticipated maturation of hiPSC-CM. In several cases, a predicted outcome was not clear because of the concerted changes in both stimulatory and inhibitory pathways. Nevertheless, clear development of hiPSC-CM over this time period was evident.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Proteoma/análise , Células Cultivadas , Fenômenos Eletrofisiológicos , Metabolismo Energético , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Mitocôndrias Cardíacas/metabolismo , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Proteoma/metabolismo , Proteômica
2.
Exp Cell Res ; 364(2): 217-223, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29458172

RESUMO

Cell transplantation has been challenged in several clinical indications of genetic or acquired muscular diseases, but therapeutic success were mitigated. To understand and improve the yields of tissue regeneration, we aimed at modelling the fate of CD56-positive human myoblasts after transplantation. Using immunodeficient severe combined immunodeficiency (SCID) mice as recipients, we assessed the survival, integration and satellite cell niche occupancy of human myoblasts by a triple immunohistochemical labelling of laminin, dystrophin and human lamin A/C. The counts were integrated into a classical mathematical decline equation. After injection, human cells were essentially located in the endomysium, then they disappeared progressively from D0 to D28. The final number of integrated human nuclei was grossly determined at D2 after injection, suggesting that no more efficient fusion between donor myoblasts and host fibers occurs after the resolution of the local damages created by needle insertion. Almost 1% of implanted human cells occupied a satellite-like cell niche. Our mathematical model validated by histological counting provided a reliable quantitative estimate of human myoblast survival and/or incorporation into SCID muscle fibers. Informations brought by histological labelling and this mathematical model are complementary.


Assuntos
Transplante de Células , Modelos Biológicos , Músculo Esquelético/citologia , Músculo Esquelético/cirurgia , Mioblastos/citologia , Mioblastos/transplante , Transplante Heterólogo , Animais , Sobrevivência Celular , Humanos , Camundongos , Camundongos Endogâmicos , Camundongos SCID , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo
3.
Stem Cell Reports ; 3(5): 905-14, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25418732

RESUMO

Cardiomyocytes from human embryonic stem cells (hESC-CMs) and induced pluripotent stem cells (hiPSC-CMs) represent new models for drug discovery. Although hypertrophy is a high-priority target, we found that hiPSC-CMs were systematically unresponsive to hypertrophic signals such as the α-adrenoceptor (αAR) agonist phenylephrine (PE) compared to hESC-CMs. We investigated signaling at multiple levels to understand the underlying mechanism of this differential responsiveness. The expression of the normal α1AR gene, ADRA1A, was reversibly silenced during differentiation, accompanied by ADRA1B upregulation in either cell type. ADRA1B signaling was intact in hESC-CMs, but not in hiPSC-CMs. We observed an increased tonic activity of inhibitory kinase pathways in hiPSC-CMs, and inhibition of antihypertrophic kinases revealed hypertrophic increases. There is tonic suppression of cell growth in hiPSC-CMs, but not hESC-CMs, limiting their use in investigation of hypertrophic signaling. These data raise questions regarding the hiPSC-CM as a valid model for certain aspects of cardiac disease.


Assuntos
Adrenérgicos/farmacologia , Tamanho Celular/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Agonistas alfa-Adrenérgicos/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Hipertrofia , Imidazóis/farmacologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Isoproterenol/farmacologia , Microscopia Confocal , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fenilefrina/farmacologia , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
4.
Mol Ther ; 21(5): 1064-75, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23439500

RESUMO

The functional and architectural benefits of embryonic stem cells (ESC) and myoblasts (Mb) transplantations into infarcted myocardium have been investigated extensively. Whereas ESC repopulated fibrotic areas and contributed to myocardial regeneration, Mb exerted their effects through paracrine secretions and scar remodeling. This therapeutic perspective, however, has been less explored in the setting of nonischemic dilated cardiomyopathies (DCMs). Our aim was to compare the integration and functional efficacy of ESC committed to cardiac fate by bone morphogenic protein 2 (BMP-2) pretreatment and Mb used as gold standard following their transplantation into the myocardium of a mouse model of laminopathy exhibiting a progressive and lethal DCM. After 4 and 8 weeks of transplantation, stabilization was observed in Mb-transplanted mice (P = 0.008) but not in groups of ESC-transplanted or medium-injected animals, where the left ventricular fractional shortening (LVFS) decreased by 32 ± 8% and 41 ± 8% respectively. Engrafted differentiated cells were consistently detected in myocardia of mice receiving Mb, whereas few or no cells were detected in the hearts of mice receiving ESC, except in two cases where teratomas were formed. These data suggest that committed ESC fail to integrate in DCM where scar tissue is absent to provide the appropriate niche, whereas the functional benefits of Mb transplantation might extend to nonischemic cardiomyopathy.


Assuntos
Cardiomiopatia Dilatada/terapia , Células-Tronco Embrionárias/transplante , Mioblastos/transplante , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/mortalidade , Cardiomiopatia Dilatada/fisiopatologia , Diferenciação Celular , Linhagem Celular , Terapia Baseada em Transplante de Células e Tecidos , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Sobrevivência de Enxerto , Masculino , Camundongos , Desenvolvimento Muscular , Mioblastos/citologia , Mioblastos/metabolismo , Contração Miocárdica , Miocárdio/metabolismo , Miocárdio/patologia , Fenótipo
5.
Curr Opin Organ Transplant ; 16(6): 640-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22001714

RESUMO

PURPOSE OF REVIEW: Cell therapy is considered a potential therapeutic avenue for the treatment of skeletal muscle diseases. Heterologous and autologous approaches have been attempted in the context, respectively, of generalized degenerative disease and of localized repairs. Cell transplantation trials, however, have been hampered by poor survival and limited migratory ability of the cells. This article reviews recent problems including the identification of new putative cellular candidates, the combination of complementary genetic or pharmacological therapeutic approaches, and the set up of clinical trials. RECENT FINDINGS: Deeper investigations identified anoikis, oxidative stress, fusion inability and some administration methodologies as causes of early massive cell death. It was proposed to adapt the injection strategies or to combine them with genetic modifications of the cells or pharmacological interventions on the environment to improve the success of implantation. New myogenic cell types have been identified, mainly in the family of perivascular cells, which can be administered systemically. New concepts have emerged regarding the correction of gene expression (use of lentiviral vectors, set-up of exon skipping, direct DNA repair, etc.). SUMMARY: Initial cell transplantation trials dedicated to the repair of striated muscles in muscular dystrophies produced mitigated results and underlined some limitations of cellular candidates under study. The research and identification of new stem cell candidates, the invention of new molecular strategies for correction of gene expression, the development of complementary approaches to improve transplantation success, have been justified by the unmet medical needs. These efforts led to new preclinical and clinical trials based on these concepts.


Assuntos
Músculo Esquelético/cirurgia , Distrofias Musculares/cirurgia , Mioblastos Esqueléticos/transplante , Transplante de Células-Tronco , Animais , Terapia Combinada , Terapia Genética , Humanos , Desenvolvimento Muscular , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofias Musculares/genética , Distrofias Musculares/patologia , Distrofias Musculares/fisiopatologia , Fármacos Neuromusculares/uso terapêutico , Recuperação de Função Fisiológica , Transplante de Células-Tronco/efeitos adversos , Resultado do Tratamento
6.
Exp Cell Res ; 316(15): 2513-26, 2010 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-20430024

RESUMO

Human skeletal muscle is an essential source of various cellular progenitors with potential therapeutic perspectives. We first used extracellular markers to identify in situ the main cell types located in a satellite position or in the endomysium of the skeletal muscle. Immunohistology revealed labeling of cells by markers of mesenchymal (CD13, CD29, CD44, CD47, CD49, CD62, CD73, CD90, CD105, CD146, and CD15 in this study), myogenic (CD56), angiogenic (CD31, CD34, CD106, CD146), hematopoietic (CD10, CD15, CD34) lineages. We then analysed cell phenotypes and fates in short- and long-term cultures of dissociated muscle biopsies in a proliferation medium favouring the expansion of myogenic cells. While CD56(+) cells grew rapidly, a population of CD15(+) cells emerged, partly from CD56(+) cells, and became individualized. Both populations expressed mesenchymal markers similar to that harboured by human bone marrow-derived mesenchymal stem cells. In differentiation media, both CD56(+) and CD15(+) cells shared osteogenic and chondrogenic abilities, while CD56(+) cells presented a myogenic capacity and CD15(+) cells presented an adipogenic capacity. An important proportion of cells expressed the CD34 antigen in situ and immediately after muscle dissociation. However, CD34 antigen did not persist in culture and this initial population gave rise to adipogenic cells. These results underline the diversity of human muscle cells, and the shared or restricted commitment abilities of the main lineages under defined conditions.


Assuntos
Células-Tronco Mesenquimais/citologia , Músculo Esquelético/citologia , Biomarcadores/análise , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Separação Celular/métodos , Células Cultivadas , Células Clonais , Expressão Gênica , Humanos , Imunofenotipagem , Hibridização in Situ Fluorescente , Magnetismo , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Microesferas , Músculo Esquelético/imunologia , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia
7.
Mol Ther ; 17(11): 1948-58, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19738599

RESUMO

Aldehyde dehydrogenase 1A1 (ALDH) activity is one hallmark of human bone marrow (BM), umbilical cord blood (UCB), and peripheral blood (PB) primitive progenitors presenting high reconstitution capacities in vivo. In this study, we have identified ALDH(+) cells within human skeletal muscles, and have analyzed their phenotypical and functional characteristics. Immunohistofluorescence analysis of human muscle tissue sections revealed rare endomysial cells. Flow cytometry analysis using the fluorescent substrate of ALDH, Aldefluor, identified brightly stained (ALDH(br)) cells with low side scatter (SSC(lo)), in enzymatically dissociated muscle biopsies, thereafter abbreviated as SMALD(+) (for skeletal muscle ALDH(+)) cells. Phenotypical analysis discriminated two sub-populations according to CD34 expression: SMALD(+)/CD34(-) and SMALD(+)/CD34(+) cells. These sub-populations did not initially express endothelial (CD31), hematopoietic (CD45), and myogenic (CD56) markers. Upon sorting, however, whereas SMALD(+)/CD34(+) cells developed in vitro as a heterogeneous population of CD56(-) cells able to differentiate in adipoblasts, the SMALD(+)/CD34(-) fraction developed in vitro as a highly enriched population of CD56(+) myoblasts able to form myotubes. Moreover, only the SMALD(+)/CD34(-) population maintained a strong myogenic potential in vivo upon intramuscular transplantation. Our results suggest that ALDH activity is a novel marker for a population of new human skeletal muscle progenitors presenting a potential for cell biology and cell therapy.


Assuntos
Aldeído Desidrogenase/metabolismo , Células Musculares/citologia , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/citologia , Adipogenia/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Fenótipo
8.
Exp Cell Res ; 313(7): 1337-46, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17362928

RESUMO

We described the ex vivo production of mature and functional human smooth muscle cells (SMCs) derived from skeletal myoblasts. Initially, myoblasts expressed all myogenic cell-related markers such as Myf5, MyoD and Myogenin and differentiate into myotubes. After culture in a medium containing vascular endothelial growth factor (VEGF), these cells were shown to have adopted a differentiated SMC identity as demonstrated by alphaSMA, SM22alpha, calponin and smooth muscle-myosin heavy chain expression. Moreover, the cells cultured in the presence of VEGF did not express MyoD anymore and were unable to fuse in multinucleated myotubes. We demonstrated that myoblasts-derived SMCs (MDSMCs) interacted with endothelial cells to form, in vitro, a capillary-like network in three-dimensional collagen culture and, in vivo, a functional vascular structure in a Matrigel implant in nonobese diabetic-severe combined immunodeficient mice. Based on the easily available tissue source and their differentiation into functional SMCs, these data argue that skeletal myoblasts might represent an important tool for SMCs-based cell therapy.


Assuntos
Diferenciação Celular , Colágeno/farmacologia , Laminina/farmacologia , Mioblastos Esqueléticos/fisiologia , Miócitos de Músculo Liso/fisiologia , Proteoglicanas/farmacologia , Técnicas de Cultura de Tecidos , Animais , Biomarcadores/análise , Células Cultivadas , Combinação de Medicamentos , Células Epiteliais/fisiologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fibras Musculares Esqueléticas/fisiologia , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/transplante , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/transplante , Neovascularização Fisiológica , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...