Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Stem Cells ; 32(5): 1059-66, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24449004

RESUMO

MicroRNAs (miRs) are highly conserved, short noncoding RNA molecules that negatively regulate messenger RNA (mRNA) stability and/or translational efficiency. Since a given miR can control the expression of many mRNAs, their importance in governing gene expression in specific cell types including vascular cells and their progenitor cells has become increasingly clear. Understanding how the expression of miRs themselves is regulated and how miRs exert their influence on post-transcriptional gene control provides novel opportunities to dissect gene regulatory networks in clinically relevant cell types. A multitude of miRs have been identified with key roles in vascular development, homeostasis, function, disease, and regeneration. In this review, we will describe the impact of miRs on angiogenesis and their capacity to modulate the behavior of stem and progenitor cells which may be utilitarian for promoting vascular growth in ischemic tissue. Moreover, we summarize these strategies available for modulating miR expression and function and future therapeutic applications.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/genética , Neovascularização Patológica/genética , Neovascularização Fisiológica/genética , Células-Tronco/metabolismo , Animais , Diferenciação Celular/genética , Células Endoteliais/metabolismo , Humanos , Modelos Genéticos , Células-Tronco/citologia
2.
Stem Cells ; 30(4): 643-54, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22232059

RESUMO

MicroRNAs (miRNAs) are short noncoding RNAs, which post-transcriptionally regulate gene expression. miRNAs are transcribed as precursors and matured to active forms by a series of enzymes, including Dicer. miRNAs are important in governing cell differentiation, development, and disease. We have recently developed a feeder- and serum-free protocol for direct derivation of endothelial cells (ECs) from human embryonic stem cells (hESCs) and provided evidence of increases in angiogenesis-associated miRNAs (miR-126 and -210) during the process. However, the functional role of miRNAs in hESC differentiation to vascular EC remains to be fully interrogated. Here, we show that the reduction of miRNA maturation induced by Dicer knockdown suppressed hES-EC differentiation. A miRNA microarray was performed to quantify hES-EC miRNA profiles during defined stages of endothelial differentiation. miR-99b, -181a, and -181b were identified as increasing in a time- and differentiation-dependent manner to peak in mature hESC-ECs and adult ECs. Augmentation of miR-99b, -181a, and -181b levels by lentiviral-mediated transfer potentiated the mRNA and protein expression of EC-specific markers, Pecam1 and VE Cadherin, increased nitric oxide production, and improved hES-EC-induced therapeutic neovascularization in vivo. Conversely, knockdown did not impact endothelial differentiation. Our results suggest that miR-99b, -181a, and -181b comprise a component of an endothelial-miRNA signature and are capable of potentiating EC differentiation from pluripotent hESCs.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/citologia , MicroRNAs/genética , Adulto , Biomarcadores/metabolismo , Linhagem Celular , Linhagem da Célula/genética , RNA Helicases DEAD-box/metabolismo , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Lentivirus/genética , MicroRNAs/metabolismo , Neovascularização Fisiológica/genética , Óxido Nítrico/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Reprodutibilidade dos Testes , Ribonuclease III/metabolismo , Transcriptoma/genética
3.
Microcirculation ; 19(3): 196-207, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22244147

RESUMO

Vascular endothelial cells derived from human pluripotent stem cells have substantial potential for the development of novel vascular therapeutics and cell-based therapies for the repair of ischemic damage. To gain maximum benefit from this source of cells, a complete understanding of the changes in gene expression and how they are regulated is required. miRNAs have been demonstrated to play a critical role in controlling stem cell pluripotency and differentiation and are important for mature endothelial cell function. Specific miRNAs that determine stem cell fate have been identified for a number of different cell lineages; however, in the case of differentiation and specification of vascular endothelial cells, this is yet to be fully elucidated.


Assuntos
MicroRNAs/genética , MicroRNAs/fisiologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Humanos , Modelos Biológicos , Neovascularização Fisiológica
4.
Arterioscler Thromb Vasc Biol ; 30(7): 1389-97, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20431067

RESUMO

OBJECTIVE: To develop an embryoid body-free directed differentiation protocol for the rapid generation of functional vascular endothelial cells derived from human embryonic stem cells (hESCs) and to assess the system for microRNA regulation and angiogenesis. METHODS AND RESULTS: The production of defined cell lineages from hESCs is a critical requirement for evaluating their potential in regenerative medicine. We developed a feeder- and serum-free protocol. Directed endothelial differentiation of hESCs revealed rapid loss of pluripotency markers and progressive induction of mRNA and protein expression of vascular markers (including CD31 and vascular endothelial [VE]-cadherin) and angiogenic growth factors (including vascular endothelial growth factor), increased expression of angiogenesis-associated microRNAs (including miR-126 and miR-210), and induction of endothelial cell morphological features. In vitro, differentiated cells produced nitric oxide, migrated across a wound, and formed tubular structures in both the absence and the presence of 3D matrices (Matrigel). In vivo, we showed that cells that differentiated for 10 days before implantation were efficient at the induction of therapeutic neovascularization and that hESC-derived cells were incorporated into the blood-perfused vasculature of recipient mice. CONCLUSIONS: The directed differentiation of hESCs is efficient and effective for the differentiation of functional endothelial cells from hESCs.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/metabolismo , Isquemia/fisiopatologia , MicroRNAs/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Cicatrização , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula , Movimento Celular , Forma Celular , Meios de Cultura Livres de Soro , Modelos Animais de Doenças , Células-Tronco Embrionárias/transplante , Células Endoteliais/transplante , Regulação da Expressão Gênica no Desenvolvimento , Membro Posterior , Humanos , Isquemia/genética , Isquemia/metabolismo , Isquemia/cirurgia , Camundongos , Neovascularização Fisiológica/genética , Óxido Nítrico/metabolismo , RNA Mensageiro/metabolismo , Transplante de Células-Tronco , Fatores de Tempo , Transfecção , Cicatrização/genética
5.
Mol Ther ; 18(12): 2139-45, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20978477

RESUMO

Retroviral vectors remain the most efficient and widely applied system for induction of pluripotency. However, mutagenic effects have been documented in both laboratory and clinical gene therapy studies, principally as a result of dysregulated host gene expression in the proximity of defined integration sites. Here, we report that cells with characteristics of pluripotent stem cells can be produced from normal human fibroblasts in the absence of reprogramming transcription factors (TFs) during lentiviral (LV) vector-mediated gene transfer. This occurred via induced alterations in host gene and microRNA (miRNA) expression and detrimental changes in karyotype. These findings demonstrate that vector-induced genotoxicity may alone play a role in somatic cell reprogramming derivation and urges caution when using integrating vectors in this setting. Clearer understanding of this process may additionally reveal novel insights into reprogramming pathways.


Assuntos
Reprogramação Celular , Fibroblastos/citologia , Lentivirus/genética , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição/genética , Perfilação da Expressão Gênica , Humanos , MicroRNAs/genética
6.
Biochem J ; 432(3): 575-84, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20923411

RESUMO

hESCs (human embryonic stem cells) have enormous potential for use in pharmaceutical development and therapeutics; however, to realize this potential, there is a requirement for simple and reproducible cell culture methods that provide adequate numbers of cells of suitable quality. We have discovered a novel way of blocking the spontaneous differentiation of hESCs in the absence of exogenous cytokines by supplementing feeder-free conditions with EHNA [erythro-9-(2-hydroxy-3-nonyl)adenine], an established inhibitor of ADA (adenosine deaminase) and cyclic nucleotide PDE2 (phosphodiesterase 2). hESCs maintained in feeder-free conditions with EHNA for more than ten passages showed no reduction in hESC-associated markers including NANOG, POU5F1 (POU domain class 5 transcription factor 1, also known as Oct-4) and SSEA4 (stage-specific embryonic antigen 4) compared with cells maintained in feeder-free conditions containing bFGF (basic fibroblast growth factor). Spontaneous differentiation was reversibly suppressed by the addition of EHNA, but, upon removing EHNA, hESC populations underwent efficient spontaneous, multi-lineage and directed differentiation. EHNA also acts as a strong blocker of directed neuronal differentiation. Chemically distinct inhibitors of ADA and PDE2 lacked the capacity of EHNA to suppress hESC differentiation, suggesting that the effect is not driven by inhibition of either ADA or PDE2. Preliminary structure-activity relationship analysis found the differentiation-blocking properties of EHNA to reside in a pharmacophore comprising a close adenine mimetic with an extended hydrophobic substituent in the 8- or 9-position. We conclude that EHNA and simple 9-alkyladenines can block directed neuronal and spontaneous differentiation in the absence of exogenous cytokine addition, and may provide a useful replacement for bFGF in large-scale or cGMP-compliant processes.


Assuntos
Adenina/análogos & derivados , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Adenina/farmacologia , Inibidores de Adenosina Desaminase/farmacologia , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Técnicas de Cultura de Células/métodos , Linhagem Celular , Células-Tronco Embrionárias/citologia , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Proteína Homeobox Nanog , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Células-Tronco Pluripotentes/citologia , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Antígenos Embrionários Estágio-Específicos/metabolismo , Relação Estrutura-Atividade , Fatores de Tempo
7.
Biochem Soc Trans ; 38(4): 1058-61, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20659003

RESUMO

hESCs (human embryonic stem cells) offer great potential for pharmaceutical research and development and, potentially, for therapeutic use. However, improvements in cell culture are urgently required to allow the scalable production of large numbers of cells that maintain pluripotency. Supplementing feeder-free conditions with either EHNA [erythro-9-(2-hydroxy-3-nonyl)adenine] or readily synthesized analogues of this compound maintains hESC pluripotency in the absence of exogenous cytokines. When the hESC lines SA121 or SA461 were maintained in feeder-free conditions with EHNA they displayed no reduction in stem-cell-associated markers such as Nanog, Oct4 (octamer-binding protein 4) and SSEA4 (stage-specific embryonic antigen 4) when compared with cells maintained in full feeder-free conditions that included exogenously added bFGF (basic fibroblast growth factor). Spontaneous differentiation was reversibly suppressed by the addition of EHNA, but EHNA did not limit efficient spontaneous or directed differentiation following its removal. We conclude that EHNA or related compounds offers a viable alternative to exogenous cytokine addition in maintaining hESC cultures in a pluripotent state and might be a particularly useful replacement for bFGF for large-scale or GMP (good manufacturing practice)-compliant processes.


Assuntos
Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/análise , Bibliotecas de Moléculas Pequenas/farmacologia , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Humanos , Ligantes , Células-Tronco Pluripotentes/fisiologia
8.
Stem Cell Res Ther ; 4(2): 36, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23618383

RESUMO

INTRODUCTION: Differentiation of vascular endothelial cells (ECs) in clinically relevant numbers for injection into ischaemic areas could offer therapeutic potential in the treatment of cardiovascular conditions, including myocardial infarction, peripheral vascular disease and stroke. While we and others have demonstrated successful generation of functional endothelial-like cells from human embryonic stem cells (hESCs), little is understood regarding the complex transcriptional and epigenetic changes that occur during differentiation, in particular during early commitment to a mesodermal lineage. METHODS: We performed the first gene expression microarray study of hESCs undergoing directed differentiation to ECs using a monolayer-based, feeder-free and serum-free protocol. Microarray results were confirmed by quantitative RT-PCR and immunocytochemistry, and chromatin immunoprecipitation (ChIP)-PCR analysis was utilised to determine the bivalent status of differentially expressed genes. RESULTS: We identified 22 transcription factors specific to early mesoderm commitment. Among these factors, FOXA2 was observed to be the most significantly differentially expressed at the hESC-EC day 2 timepoint. ChIP-PCR analysis revealed that the FOXA2 transcription start site is bivalently marked with histone modifications for both gene activation (H3K4me3) and repression (H3K27me3) in hESCs, suggesting the transcription factor may be a key regulator of hESC differentiation. CONCLUSION: This enhanced knowledge of the lineage commitment process will help improve the design of directed differentiation protocols, increasing the yield of endothelial-like cells for regenerative medicine therapies in cardiovascular disease.


Assuntos
Células Endoteliais/citologia , Perfilação da Expressão Gênica , Fator 3-beta Nuclear de Hepatócito/metabolismo , Mesoderma/metabolismo , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Células-Tronco Embrionárias/citologia , Epigênese Genética , Fator 3-beta Nuclear de Hepatócito/genética , Histonas/metabolismo , Humanos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
9.
Hum Gene Ther ; 23(12): 1247-57, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22931362

RESUMO

We have previously shown that injury-induced neointima formation was rescued by adenoviral-Nogo-B gene delivery. Integrase-competent lentiviral vectors (ICLV) are efficient at gene delivery to vascular cells but present a risk of insertional mutagenesis. Conversely, integrase-deficient lentiviral vectors (IDLV) offer additional benefits through reduced mutagenesis risk, but this has not been evaluated in the context of vascular gene transfer. Here, we have investigated the performance and genetic safety of both counterparts in primary human vascular smooth muscle cells (VSMC) and compared gene transfer efficiency and assessed the genotoxic potential of ICLVs and IDLVs based on their integration frequency and insertional profile in the human genome. Expression of enhanced green fluorescent protein (eGFP) mediated by IDLVs (IDLV-eGFP) demonstrated efficient transgene expression in VSMCs. IDLV gene transfer of Nogo-B mediated efficient overexpression of Nogo-B in VSMCs, leading to phenotypic effects on VSMC migration and proliferation, similar to its ICLV version and unlike its eGFP control and uninfected VSMCs. Large-scale integration site analyses in VSMCs indicated that IDLV-mediated gene transfer gave rise to a very low frequency of genomic integration compared to ICLVs, revealing a close-to-random genomic distribution in VSMCs. This study demonstrates for the first time the potential of IDLVs for safe and efficient vascular gene transfer.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos/genética , Integrases/genética , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Proteínas de Fluorescência Verde/genética , Humanos , Lentivirus/genética , Mutagênese Insercional , Proteínas da Mielina/genética , Proteínas Nogo
10.
Vascul Pharmacol ; 55(4): 69-78, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21854874

RESUMO

Human embryonic stem cells (hESC) offer broad potential for regenerative medicine owing to their capacity for self renewal, exponential scale up and differentiation into any cell type in the adult body. hESC have been proposed as a potentially unlimited source for the generation of transplantable, healthy, functional vascular cells for repair of ischemic tissues. To optimally harness this potential necessitates precise control over biological processes that govern maintenance, pluripotency and cell differentiation including signalling cascades, gene expression profiles and epigenetic modification. Such control may be elicited by microRNAs, which are powerful negative regulators of gene expression. Here, we review the role for miRNAs in both the maintenance of pluripotency and differentiation of cells to a cardiovascular lineage including endothelial cells, vascular smooth muscle cells and cardiomyocytes and put this into context for regenerative medicine in the cardiovascular system.


Assuntos
Moduladores da Angiogênese/metabolismo , MicroRNAs/fisiologia , Neovascularização Fisiológica , Células-Tronco Pluripotentes/metabolismo , Moduladores da Angiogênese/agonistas , Moduladores da Angiogênese/antagonistas & inibidores , Animais , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica , Humanos , MicroRNAs/agonistas , MicroRNAs/antagonistas & inibidores , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Neovascularização Patológica/metabolismo , Células-Tronco Pluripotentes/citologia , Medicina Regenerativa/métodos
11.
Pharmacol Ther ; 129(1): 29-49, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20965210

RESUMO

Several types of stem and progenitor cells are currently under investigation for their potential to accomplish vascular regeneration. This review focuses on embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). We will discuss the technologies allowing for their derivation, culture expansion and maintenance in a pluripotent status. Moreover, both ESCs and iPSCs can be differentiated in endothelial cells (ECs) and mural cell, including vascular smooth muscle cells (VSMCs). Here, we will describe the involvements of growth factors (vascular endothelial growth factors-VEGFs-, platet-derived growth factors-PDGFs-), Wnt and Notch signal pathways, reactive oxygen species (ROS), histone deacetylases (HDACs), and microRNAs (miRNAs) in vascular cell differentiation from pluripotent stem cells. We will additionally describe the therapeutic potential of stem cells for vascular medicine.


Assuntos
Vasos Sanguíneos/citologia , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células Endoteliais/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos de Músculo Liso/citologia , Vasos Sanguíneos/fisiologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/transplante , Células Endoteliais/fisiologia , Histona Desacetilases/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , MicroRNAs/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Regeneração , Transdução de Sinais
12.
PLoS One ; 5(9): e12970, 2010 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-20885978

RESUMO

BACKGROUND: Decidualization (differentiation) of the endometrial stromal cells during the secretory phase of the menstrual cycle is essential for successful implantation. Transforming Growth Factor ß1 (TGFß1) canonically propagates its actions via SMAD signalling. A role for TGFß1 in decidualization remains to be established and published data concerning effects of TGFß1 on markers of endometrial decidualization are inconsistent. METHODOLOGY/PRINCIPAL FINDINGS: Non-pregnant endometrial stromal cells (ESC) and first trimester decidual stromal cells (DSC) were cultured in the presence or absence of a decidualizing stimulus. Incubation of ESCs with TGFß1 (10 ng/ml) down-regulated the expression of transcripts encoding the decidual marker proteins prolactin (PRL), insulin-like growth factor binding protein-1 (IGFBP-1) and tissue factor (TF). TGFß1 also inhibited secretion of PRL and IGFBP-1 proteins by ESCs and surprisingly this response preceded down-regulation of their mRNAs. In contrast, DSCs were more refractory to the actions of TGFß1, characterized by blunted and delayed down-regulation of PRL, IGFBP-1, and TF transcripts, which was not associated with a significant reduction in secretion of PRL or IGFBP-1 proteins. Addition of an antibody directed against TGFß1 increased expression of IGFBP-1 mRNA in decidualised cells. Knockdown of SMAD 4 using siRNAs abrogated the effect of TGFß1 on expression of PRL in ESCs but did not fully restore expression of IGFBP-1 mRNA and protein. CONCLUSIONS/SIGNIFICANCE: TGFß1 inhibits the expression and secretion of decidual marker proteins. The impact of TGFß1 on PRL is SMAD-dependent but the impact on IGFBP1 is via an alternative mechanism. In early pregnancy, resistance of DSC to the impact of TGFß1 may be important to ensure tissue homeostasis.


Assuntos
Endométrio/metabolismo , Regulação da Expressão Gênica , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Prolactina/genética , Proteína Smad4/metabolismo , Células Estromais/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Diferenciação Celular , Células Cultivadas , Regulação para Baixo , Endométrio/citologia , Feminino , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Gravidez , Prolactina/metabolismo , Transdução de Sinais , Proteína Smad4/genética , Células Estromais/citologia , Fator de Crescimento Transformador beta1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA