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.
Sci Rep ; 7(1): 10075, 2017 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-28855627

RESUMO

Induced pluripotent stem cell-derived neural stem cells (iNSCs) have significant potential as an autologous, multifunctional cell therapy for stroke, which is the primary cause of long term disability in the United States and the second leading cause of death worldwide. Here we show that iNSC transplantation improves recovery through neuroprotective, regenerative, and cell replacement mechanisms in a novel ischemic pig stroke model. Longitudinal multiparametric magnetic resonance imaging (MRI) following iNSC therapy demonstrated reduced changes in white matter integrity, cerebral blood perfusion, and brain metabolism in the infarcted tissue. The observed tissue level recovery strongly correlated with decreased immune response, enhanced neuronal protection, and increased neurogenesis. iNSCs differentiated into neurons and oligodendrocytes with indication of long term integration. The robust recovery response to iNSC therapy in a translational pig stroke model with increased predictive potential strongly supports that iNSCs may be the critically needed therapeutic for human stroke patients.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Infarto da Artéria Cerebral Média/terapia , Transplante de Células-Tronco/métodos , Acidente Vascular Cerebral/terapia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Infarto da Artéria Cerebral Média/diagnóstico por imagem , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Imageamento por Ressonância Magnética/métodos , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Neurônios/patologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia , Suínos , Substância Branca/diagnóstico por imagem , Substância Branca/metabolismo , Substância Branca/patologia
2.
Cell Transplant ; 22(6): 945-59, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23043799

RESUMO

Neural cells derived from induced pluripotent stem cells (iPSCs) have the potential for autologous cell therapies in treating patients with severe neurological disorders or injury. However, further study of efficacy and safety are needed in large animal preclinical models that have similar neural anatomy and physiology to humans such as the pig. The pig model for pluripotent stem cell therapy has been made possible for the first time with the development of pig iPSCs (piPSCs) capable of in vitro and in vivo differentiation into tissues of all three germ layers. Still, the question remains if piPSCs are capable of undergoing robust neural differentiation using a system similar to those being used with human iPSCs. In this study, we generated a new line of piPSCs from fibroblast cells that expressed pluripotency markers and were capable of embryoid body differentiation into all three germ layers. piPSCs demonstrated robust neural differentiation forming ßIII-TUB/MAP2+ neurons, GFAP+ astrocytes, and O4+ oligodendrocytes and demonstrated strong upregulation of neural cell genes representative of all three major neural lineages of the central nervous system. In the presence of motor neuron signaling factors, piPSC-derived neurons showed expression of transcription factors associated with motor neuron differentiation (HB9 and ISLET1). Our findings demonstrate that SSEA4 expression is required for piPSCs to differentiate into neurons, astrocytes, and oligodendrocytes and furthermore develop specific neuronal subtypes. This indicates that the pigs can fill the need for a powerful model to study autologous neural iPSC therapies in a system similar to humans.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/citologia , Antígenos Embrionários Estágio-Específicos/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Biomarcadores/metabolismo , Forma Celular , Reprogramação Celular/genética , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Humanos , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Neurônios/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Sus scrofa , Transdução Genética , Tubulina (Proteína)/metabolismo
3.
Biotechnol Bioeng ; 109(1): 274-83, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21837664

RESUMO

Human mesenchymal stem cells (hMSC) have proven beneficial in the repair and preservation of infarcted myocardium. Unfortunately, MSCs represent a small portion of the bone marrow and require ex vivo expansion. To further advance the clinical usefulness of cellular cardiomyoplasty, derivation of "MSC-like" cells that can be made available "off-the-shelf" are desirable. Recently, human embryonic stem cell-derived mesenchymal cells (hESC-MC) were described. We investigated the efficacy of hESC-MC for cardiac repair after myocardial infarction (MI) compared to hMSC. Because of increased efficacy of cell delivery, cells were embedded into collagen patches and delivered to infarcted myocardium. Culture of hMSC and hESC-MCs in collagen patches did not induce differentiation or significant loss in viability. Transplantation of hMSC and hES-MC patches onto infarcted myocardium of athymic nude rats prevented adverse changes in infarct wall thickness and fractional area change compared to a non-viable patch control. Hemodynamic assessment showed that hMSCs and hES-MC patch application improved end diastolic pressure equivalently. There were no changes in systolic function. hES-MC and hMSC construct application enhanced neovessel formation compared to a non-viable control, and each cell type had similar efficacy in stimulating endothelial cell growth in vitro. In summary, the use of hES-MC provides similar efficacy for cellular cardiomyoplasty as compared to hMSC and may be considered a suitable alternative for cell therapy.


Assuntos
Células-Tronco Embrionárias/fisiologia , Infarto do Miocárdio/terapia , Engenharia Tecidual/métodos , Animais , Pressão Sanguínea/fisiologia , Diferenciação Celular , Sobrevivência Celular , Modelos Animais de Doenças , Humanos , Células-Tronco Mesenquimais/fisiologia , Ratos , Resultado do Tratamento
4.
Pharmacol Ther ; 129(3): 290-306, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21073897

RESUMO

Stem cell-based therapeutics have the potential to effectively treat many terminal and debilitating human diseases, but the mechanisms by which their growth and differentiation are regulated are incompletely defined. Recent data from multiple systems suggest major roles for G protein coupled receptor (GPCR) pathways in regulating stem cell function in vivo and in vitro. The goal of this review is to illustrate common ground between the growing field of stem cell therapeutics and the long-established field of G protein coupled receptor signaling. Herein, we briefly introduce basic stem cell biology and discuss how several conserved pathways regulate pluripotency and differentiation in mouse and human stem cells. We further discuss general mechanisms by which GPCR signaling may impact these pluripotency and differentiation pathways, and summarize specific examples of receptors from each of the major GPCR subfamilies that have been shown to regulate stem cell function. Finally, we discuss possible therapeutic implications of GPCR regulation of stem cell function.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Pluripotentes/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Humanos , Células-Tronco Pluripotentes/citologia , Receptores Acoplados a Proteínas G/metabolismo
5.
Microsc Microanal ; 16(1): 80-90, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20082731

RESUMO

Neurodegerative disorders affect millions of people worldwide. Neural cells derived from human embryonic stem cells (hESC) have the potential for cell therapies and/or compound screening for treating affected individuals. While both protein and gene expression indicative of a neural phenotype has been exhibited in these differentiated cells, ultrastuctural studies thus far have been lacking. The objective of this study was to correlate hESC to neural differentiation culture conditions with ultrastructural changes observed in the treated cells. We demonstrate here that in basic culture conditions without growth factors or serum we obtain neural morphology. The addition of brain-derived neurotrophic factor (BDNF) and serum to cultures resulted in more robust neural differentiation. In addition to providing cues such as cell survival or lineage specification, additional factors also altered the intracellular structures and cell morphologies. Even though the addition of BDNF and serum did not increase synaptic formation, altered cellular structures such as abundant polyribosomes and more developed endoplasmic reticulum indicate a potential increase in protein production.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/ultraestrutura , Neurônios/fisiologia , Neurônios/ultraestrutura , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Células Cultivadas , Meios de Cultura/química , Substâncias de Crescimento/farmacologia , Humanos
6.
Stem Cells Dev ; 19(8): 1257-66, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19886822

RESUMO

Mesenchymal stem cells (MSCs) can differentiate into multiple mesodermal cell types in vitro; however, their differentiation capacity is influenced by their tissue of origin. To what extent epigenetic information on promoters of lineage-specification genes in human progenitors influences transcriptional activation and differentiation potential remains unclear. We produced bisulfite sequencing maps of DNA methylation in adipogenic, myogenic, and endothelial promoters in relation to gene expression and differentiation capacity, and unravel a similarity in DNA methylation profiles between MSCs isolated from human adipose tissue, bone marrow (BM), and muscle. This similarity is irrespective of promoter CpG content. Methylation patterns of MSCs are distinct from those of hematopoietic progenitor cells (HPCs), pluripotent human embryonic stem cells (hESCs), and multipotent hESC-derived mesenchymal cells (MCs). Moreover, in vitro MSC differentiation does not affect lineage-specific promoter methylation states, arguing that these methylation patterns in differentiated cells are already established at the progenitor stage. Further, we find a correlation between lineage-specific promoter hypermethylation and lack of differentiation capacity toward that lineage, but no relationship between weak promoter methylation and capacity of transcriptional activation or differentiation. Thus, only part of the restriction in differentiation capacity of tissue-specific stem cells is programmed by promoter DNA methylation: hypermethylation seems to constitute a barrier to differentiation, however, no or weak methylation has no predictive value for differentiation potential.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Linhagem da Célula/fisiologia , Metilação de DNA , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Regiões Promotoras Genéticas/genética , Adipócitos/citologia , Adipócitos/metabolismo , Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Ilhas de CpG/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Expressão Gênica/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células Musculares/citologia , Células Musculares/metabolismo , Músculo Esquelético/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética
7.
BMC Neurosci ; 9: 118, 2008 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19077254

RESUMO

BACKGROUND: Lysophospholipids regulate the morphology and growth of neurons, neural cell lines, and neural progenitors. A stable human neural progenitor cell line is not currently available in which to study the role of lysophospholipids in human neural development. We recently established a stable, adherent human embryonic stem cell-derived neuroepithelial (hES-NEP) cell line which recapitulates morphological and phenotypic features of neural progenitor cells isolated from fetal tissue. The goal of this study was to determine if hES-NEP cells express functional lysophospholipid receptors, and if activation of these receptors mediates cellular responses critical for neural development. RESULTS: Our results demonstrate that Lysophosphatidic Acid (LPA) and Sphingosine-1-phosphate (S1P) receptors are functionally expressed in hES-NEP cells and are coupled to multiple cellular signaling pathways. We have shown that transcript levels for S1P1 receptor increased significantly in the transition from embryonic stem cell to hES-NEP. hES-NEP cells express LPA and S1P receptors coupled to G i/o G-proteins that inhibit adenylyl cyclase and to G q-like phospholipase C activity. LPA and S1P also induce p44/42 ERK MAP kinase phosphorylation in these cells and stimulate cell proliferation via G i/o coupled receptors in an Epidermal Growth Factor Receptor (EGFR)- and ERK-dependent pathway. In contrast, LPA and S1P stimulate transient cell rounding and aggregation that is independent of EGFR and ERK, but dependent on the Rho effector p160 ROCK. CONCLUSION: Thus, lysophospholipids regulate neural progenitor growth and morphology through distinct mechanisms. These findings establish human ES cell-derived NEP cells as a model system for studying the role of lysophospholipids in neural progenitors.


Assuntos
Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Células Neuroepiteliais/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Proliferação de Células , Células-Tronco Embrionárias/citologia , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Humanos , Microscopia de Vídeo , Células Neuroepiteliais/citologia , Células Neuroepiteliais/fisiologia , RNA Mensageiro/metabolismo , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/fisiologia , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Quinases Associadas a rho/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...