Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Circulation ; 148(14): 1099-1112, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37602409

RESUMO

BACKGROUND: Cardiac reprogramming is a technique to directly convert nonmyocytes into myocardial cells using genes or small molecules. This intervention provides functional benefit to the rodent heart when delivered at the time of myocardial infarction or activated transgenically up to 4 weeks after myocardial infarction. Yet, several hurdles have prevented the advancement of cardiac reprogramming for clinical use. METHODS: Through a combination of screening and rational design, we identified a cardiac reprogramming cocktail that can be encoded in a single adeno-associated virus. We also created a novel adeno-associated virus capsid that can transduce cardiac fibroblasts more efficiently than available parental serotypes by mutating posttranslationally modified capsid residues. Because a constitutive promoter was needed to drive high expression of these cell fate-altering reprogramming factors, we included binding sites to a cardiomyocyte-restricted microRNA within the 3' untranslated region of the expression cassette that limits expression to nonmyocytes. After optimizing this expression cassette to reprogram human cardiac fibroblasts into induced cardiomyocyte-like cells in vitro, we also tested the ability of this capsid/cassette combination to confer functional benefit in acute mouse myocardial infarction and chronic rat myocardial infarction models. RESULTS: We demonstrated sustained, dose-dependent improvement in cardiac function when treating a rat model 2 weeks after myocardial infarction, showing that cardiac reprogramming, when delivered in a single, clinically relevant adeno-associated virus vector, can support functional improvement in the postremodeled heart. This benefit was not observed with GFP (green fluorescent protein) or a hepatocyte reprogramming cocktail and was achieved even in the presence of immunosuppression, supporting myocyte formation as the underlying mechanism. CONCLUSIONS: Collectively, these results advance the application of cardiac reprogramming gene therapy as a viable therapeutic approach to treat chronic heart failure resulting from ischemic injury.


Assuntos
MicroRNAs , Infarto do Miocárdio , Ratos , Camundongos , Humanos , Animais , Dependovirus/genética , Miócitos Cardíacos/metabolismo , Infarto do Miocárdio/terapia , Infarto do Miocárdio/tratamento farmacológico , MicroRNAs/genética , MicroRNAs/metabolismo , Terapia Genética/métodos , Proteínas de Fluorescência Verde/genética , Reprogramação Celular , Fibroblastos/metabolismo
2.
J Clin Ultrasound ; 50(9): 1368-1372, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36149806

RESUMO

Vanishing bile duct syndrome (VBDS) is a pathological concept that refers to the gradual reduction, destruction and disappearance of intrahepatic bile ducts caused by drugs, immunity, malignancy, and infections (including HIV and tuberculosis). Its clinical manifestation is cholestasis. The pathological diagnostic criteria for VBDS are the occurrence of intralobular vanishing bile ducts in more than 50% of 10 or more portal areas. At present, the diagnosis of VBDS still relies on liver biopsy. Contrast-enhanced ultrasound has been widely used in the diagnosis of liver-related diseases. The intravenous injection of microbubbles could enhance the observation of tissue microcirculation and significantly expand the possibility of ultrasound hemodynamic research. VBDS is a rare disease, and there are few reports on the early ultrasound and CEUS manifestations. The purpose of this report is to explore the unique performance of ultrasound and CEUS in the diagnosis of VBDS.


Assuntos
Colestase , Cirrose Hepática Biliar , Humanos , Cirrose Hepática Biliar/complicações , Cirrose Hepática Biliar/patologia , Colestase/diagnóstico , Colestase/etiologia , Colestase/patologia , Ductos Biliares Intra-Hepáticos/patologia , Ductos Biliares , Ultrassonografia/efeitos adversos
3.
Int J Gen Med ; 15: 5159-5171, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35642202

RESUMO

Background: At present, there is a lack of cheap, effective and convenient detection methods for hepatitis B-related liver fibrosis, especially in the developing area. Aim: To evaluate the non-invasive methods for the significant and advanced fibrosis stage in chronic hepatitis B virus (HBV) patients in basic hospitals and to assess their diagnostic utility. Methods: The study included 436 consecutive naive HBV individuals who had their livers biopsied. They were examined in one week using aspartate aminotransferase-to-aspartate aminotransferase ratio (AAR), age-platelet index (API), aspartate aminotransferase-to-platelet ratio index (APRI), fibrosis-4 (FIB-4), Forns, gamma-glutamyl transpeptidase-to-platelet ratio (GPR), S-index and transient elastography (TE). Scheuer scoring system was used to determine the histologic fibrosis grades (S0-S4). The diagnostic effectiveness was assessed using AUROCs and the DeLong test, both of which were based on statistical comparisons. Results: For both substantial (≧S2) and advanced (≧S3) fibrosis phases, TE had good diagnostic performance in determining the hepatic fibrosis. Similar diagnostic performance was shown with Forns and S-index when it came to detecting fibrosis stages lower than S3. One model's diagnostic value was not significantly improved by combining serum models. Correlation coefficients between clinical features and fibrosis phases were greatest for Forns (r = 0.397), S-index (r = 0.382) and TE (r = 0.535) when compared to other variables. Conclusion: This investigation showed that Forns and S-index may be helpful strategies for detecting advanced fibrosis in HBV patients admitted to community hospitals.

4.
Cell Stem Cell ; 25(1): 87-102.e9, 2019 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-31271750

RESUMO

Ectopic expression of combinations of transcription factors (TFs) can drive direct lineage conversion, thereby reprogramming a somatic cell's identity. To determine the molecular mechanisms by which Gata4, Mef2c, and Tbx5 (GMT) induce conversion from a cardiac fibroblast toward an induced cardiomyocyte, we performed comprehensive transcriptomic, DNA-occupancy, and epigenomic interrogation throughout the reprogramming process. Integration of these datasets identified new TFs involved in cardiac reprogramming and revealed context-specific roles for GMT, including the ability of Mef2c and Tbx5 to independently promote chromatin remodeling at previously inaccessible sites. We also find evidence for cooperative facilitation and refinement of each TF's binding profile in a combinatorial setting. A reporter assay employing newly defined regulatory elements confirmed that binding of a single TF can be sufficient for gene activation, suggesting that co-binding events do not necessarily reflect synergy. These results shed light on fundamental mechanisms by which combinations of TFs direct lineage conversion.


Assuntos
Fator de Transcrição GATA4/metabolismo , Fatores de Transcrição MEF2/metabolismo , Miócitos Cardíacos/fisiologia , Proteínas com Domínio T/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Reprogramação Celular , Montagem e Desmontagem da Cromatina , Epigênese Genética , Fator de Transcrição GATA4/genética , Fatores de Transcrição MEF2/genética , Aprendizado de Máquina , Camundongos , Ligação Proteica , Proteínas com Domínio T/genética , Ativação Transcricional
5.
Circulation ; 135(10): 978-995, 2017 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-27834668

RESUMO

BACKGROUND: Reprogramming of cardiac fibroblasts into induced cardiomyocyte-like cells in situ represents a promising strategy for cardiac regeneration. A combination of 3 cardiac transcription factors, Gata4, Mef2c, and Tbx5 (GMT), can convert fibroblasts into induced cardiomyocyte-like cells, albeit with low efficiency in vitro. METHODS: We screened 5500 compounds in primary cardiac fibroblasts to identify the pathways that can be modulated to enhance cardiomyocyte reprogramming. RESULTS: We found that a combination of the transforming growth factor-ß inhibitor SB431542 and the WNT inhibitor XAV939 increased reprogramming efficiency 8-fold when added to GMT-overexpressing cardiac fibroblasts. The small molecules also enhanced the speed and quality of cell conversion; we observed beating cells as early as 1 week after reprogramming compared with 6 to 8 weeks with GMT alone. In vivo, mice exposed to GMT, SB431542, and XAV939 for 2 weeks after myocardial infarction showed significantly improved reprogramming and cardiac function compared with those exposed to only GMT. Human cardiac reprogramming was similarly enhanced on transforming growth factor-ß and WNT inhibition and was achieved most efficiently with GMT plus myocardin. CONCLUSIONS: Transforming growth factor-ß and WNT inhibitors jointly enhance GMT-induced direct cardiac reprogramming from cardiac fibroblasts in vitro and in vivo and provide a more robust platform for cardiac regeneration.


Assuntos
Benzamidas/farmacologia , Reprogramação Celular/efeitos dos fármacos , Dioxóis/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Fatores de Transcrição/metabolismo , Animais , Benzamidas/uso terapêutico , Células Cultivadas , Dioxóis/uso terapêutico , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Coração/diagnóstico por imagem , Compostos Heterocíclicos com 3 Anéis/uso terapêutico , Humanos , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Imageamento por Ressonância Magnética , Camundongos , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/metabolismo
6.
Biomaterials ; 103: 1-11, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27376554

RESUMO

Reprogramming of fibroblasts to cardiomyocytes offers exciting potential in cell therapy and regenerative medicine, but has low efficiency. We hypothesize that physical cues may positively affect the reprogramming process, and studied the effects of periodic mechanical stretch, substrate stiffness and microgrooved substrate on reprogramming yield. Subjecting reprogramming fibroblasts to periodic mechanical stretch and different substrate stiffness did not improve reprogramming yield. On the other hand, culturing the cells on microgrooved substrate enhanced the expression of cardiomyocyte genes by day 2 and improved the yield of partially reprogrammed cells at day 10. By combining microgrooved substrate with an existing optimized culture protocol, yield of reprogrammed cardiomyocytes with striated cardiac troponin T staining and spontaneous contractile activity was increased. We identified the regulation of Mkl1 activity as a new mechanism by which microgroove can affect reprogramming. Biochemical approach could only partially recapitulate the effect of microgroove. Microgroove demonstrated an additional effect of enhancing organization of sarcomeric structure, which could not be recapitulated by biochemical approach. This study provides insights into new mechanisms by which topographical cues can affect cellular reprogramming.


Assuntos
Reprogramação Celular/fisiologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Mecanotransdução Celular/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Alicerces Teciduais , Animais , Animais Recém-Nascidos , Técnicas de Cultura Celular por Lotes/métodos , Células Cultivadas , Módulo de Elasticidade/fisiologia , Camundongos , Camundongos Transgênicos , Resistência à Tração/fisiologia , Engenharia Tecidual/métodos
7.
Int J Nanomedicine ; 10: 1841-54, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25834424

RESUMO

The treatment of myocardial infarction is a major challenge in medicine due to the inability of heart tissue to regenerate. Direct reprogramming of endogenous cardiac fibroblasts into functional cardiomyocytes via the delivery of transcription factor mRNAs has the potential to regenerate cardiac tissue and to treat heart failure. Even though mRNA delivery to cardiac fibroblasts has the therapeutic potential, mRNA transfection in cardiac fibroblasts has been challenging. Herein, we develop an efficient mRNA transfection in cultured mouse cardiac fibroblasts via a polyarginine-fused heart-targeting peptide and lipofectamine complex, termed C-Lipo and demonstrate the partial direct reprogramming of cardiac fibroblasts towards cardiomyocyte cells. C-Lipo enabled the mRNA-induced direct cardiac reprogramming due to its efficient transfection with low toxicity, which allowed for multiple transfections of Gata4, Mef2c, and Tbx5 (GMT) mRNAs for a period of 2 weeks. The induced cardiomyocyte-like cells had α-MHC promoter-driven GFP expression and striated cardiac muscle structure from α-actinin immunohistochemistry. GMT mRNA transfection of cultured mouse cardiac fibroblasts via C-Lipo significantly increased expression of the cardiomyocyte marker genes, Actc1, Actn2, Gja1, Hand2, and Tnnt2, after 2 weeks of transfection. Moreover, this study provides the first direct evidence that the stoichiometry of the GMT reprogramming factors influence the expression of cardiomyocyte marker genes. Our results demonstrate that mRNA delivery is a potential approach for cardiomyocyte generation.


Assuntos
Reprogramação Celular/fisiologia , Fibroblastos/fisiologia , Miócitos Cardíacos/citologia , RNA Mensageiro/genética , Transfecção/métodos , Animais , Células Cultivadas , Fibroblastos/citologia , Regulação da Expressão Gênica , Marcadores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Lipídeos/administração & dosagem , Lipídeos/química , Lipídeos/toxicidade , Camundongos Transgênicos , Miocárdio/citologia , Miocárdio/ultraestrutura , Miócitos Cardíacos/fisiologia , Oligopeptídeos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas com Domínio T/genética
8.
Stem Cell Reports ; 2(1): 18-25, 2014 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-24678451

RESUMO

NANOG is a divergent homeobox protein and a core component of the transcriptional circuitry that sustains pluripotency and self-renewal. Although NANOG has been extensively studied on the transcriptional level, little is known regarding its posttranslational regulation, likely due to its low abundance and challenging physical properties. Here, we identify eleven phosphorylation sites on endogenous human NANOG, nine of which mapped to single amino acids. To screen for the signaling molecules that impart these modifications, we developed the multiplexed assay for kinase specificity (MAKS). MAKS simultaneously tests activity for up to ten kinases while directly identifying the substrate and exact site of phosphorylation. Using MAKS, we discovered site-specific phosphorylation by ERK2 and CDK1/CyclinA2, providing a putative link between key signaling pathways and NANOG.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Sequência de Aminoácidos , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Ciclina A2/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/isolamento & purificação , Humanos , Dados de Sequência Molecular , Proteína Homeobox Nanog , Fosfopeptídeos/análise , Fosforilação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
9.
Cell ; 153(5): 1134-48, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23664764

RESUMO

Epigenetic mechanisms have been proposed to play crucial roles in mammalian development, but their precise functions are only partially understood. To investigate epigenetic regulation of embryonic development, we differentiated human embryonic stem cells into mesendoderm, neural progenitor cells, trophoblast-like cells, and mesenchymal stem cells and systematically characterized DNA methylation, chromatin modifications, and the transcriptome in each lineage. We found that promoters that are active in early developmental stages tend to be CG rich and mainly engage H3K27me3 upon silencing in nonexpressing lineages. By contrast, promoters for genes expressed preferentially at later stages are often CG poor and primarily employ DNA methylation upon repression. Interestingly, the early developmental regulatory genes are often located in large genomic domains that are generally devoid of DNA methylation in most lineages, which we termed DNA methylation valleys (DMVs). Our results suggest that distinct epigenetic mechanisms regulate early and late stages of ES cell differentiation.


Assuntos
Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Epigenômica , Regulação da Expressão Gênica no Desenvolvimento , Animais , Diferenciação Celular , Cromatina/metabolismo , Ilhas de CpG , Células-Tronco Embrionárias/citologia , Histonas/metabolismo , Humanos , Metilação , Neoplasias/genética , Regiões Promotoras Genéticas , Peixe-Zebra/embriologia
10.
Proc Natl Acad Sci U S A ; 109(22): 8411-6, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22586074

RESUMO

We have developed and implemented a sequence identification algorithm (inSeq) that processes tandem mass spectra in real-time using the mass spectrometer's (MS) onboard processors. The inSeq algorithm relies on accurate mass tandem MS data for swift spectral matching with high accuracy. The instant spectral processing technology takes ∼16 ms to execute and provides information to enable autonomous, real-time decision making by the MS system. Using inSeq and its advanced decision tree logic, we demonstrate (i) real-time prediction of peptide elution windows en masse (∼3 min width, 3,000 targets), (ii) significant improvement of quantitative precision and accuracy (~3x boost in detected protein differences), and (iii) boosted rates of posttranslation modification site localization (90% agreement in real-time vs. offline localization rate and an approximate 25% gain in localized sites). The decision tree logic enabled by inSeq promises to circumvent problems with the conventional data-dependent acquisition paradigm and provides a direct route to streamlined and expedient targeted protein analysis.


Assuntos
Algoritmos , Peptídeos/análise , Proteínas/análise , Espectrometria de Massas em Tandem/métodos , Sequência de Aminoácidos , Sítios de Ligação , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Bases de Dados de Proteínas , Árvores de Decisões , Humanos , Dados de Sequência Molecular , Peptídeos/química , Processamento de Proteína Pós-Traducional , Proteínas/química , Reprodutibilidade dos Testes , Software , Fatores de Tempo
11.
Proc Natl Acad Sci U S A ; 109(19): 7162-8, 2012 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-22474382

RESUMO

The transcription factor OCT4 is fundamental to maintaining pluripotency and self-renewal. To better understand protein-level regulation of OCT4, we applied liquid chromatography-MS to identify 14 localized sites of phosphorylation, 11 of which were previously unknown. Functional analysis of two sites, T234 and S235, suggested that phosphorylation within the homeobox region of OCT4 negatively regulates its activity by interrupting sequence-specific DNA binding. Mutating T234 and S235 to mimic constitutive phosphorylation at these sites reduces transcriptional activation from an OCT4-responsive reporter and decreases reprogramming efficiency. We also cataloged 144 unique phosphopeptides on known OCT4 interacting partners, including SOX2 and SALL4, that copurified during immunoprecipitation. These proteins were enriched for phosphorylation at motifs associated with ERK signaling. Likewise, OCT4 harbored several putative ERK phosphorylation sites. Kinase assays confirmed that ERK2 phosphorylated these sites in vitro, providing a direct link between ERK signaling and the transcriptional machinery that governs pluripotency.


Assuntos
Células-Tronco Embrionárias/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Serina/metabolismo , Treonina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/genética , Western Blotting , Células Cultivadas , Células HEK293 , Humanos , Imunoprecipitação , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Fator 3 de Transcrição de Octâmero/química , Fator 3 de Transcrição de Octâmero/genética , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Fatores de Transcrição SOXB1/metabolismo , Homologia de Sequência de Aminoácidos , Serina/química , Serina/genética , Treonina/química , Treonina/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional
12.
Stem Cells ; 30(4): 623-30, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22213113

RESUMO

Fibroblast growth factor (FGF), transforming growth factor (TGF)/Nodal, and Insulin/insulin-like growth factor (IGF) signaling pathways are sufficient to maintain human embryonic stem cells (ESCs) and induced pluripotent stem cells in a proliferative, undifferentiated state. Here, we show that only a few FGF family members (FGF2, FGF4, FGF6, and FGF9) are able to sustain strong extracellular-signal-regulated kinase (ERK) phosphorylation and NANOG expression levels in human ESCs. Surprisingly, FGF1, which is reported to target the same set of receptors as FGF2, fails to sustain ERK phosphorylation and NANOG expression under standard culture conditions. We find that the failure of FGF1 to sustain ES is due to thermal instability of the wild-type protein, not receptor specificity, and that a mutated thermal-stable FGF1 sustains human ESCs and supports both differentiation and reprogramming protocols.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/farmacologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Temperatura , Proliferação de Células/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Heparina/metabolismo , Humanos , Proteínas Mutantes/metabolismo , Mutação/genética , Fosforilação/efeitos dos fármacos , Células-Tronco Pluripotentes/enzimologia , Estabilidade Proteica/efeitos dos fármacos , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo
13.
Cell Res ; 21(10): 1393-409, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21876557

RESUMO

Pluripotency, the ability of a cell to differentiate and give rise to all embryonic lineages, defines a small number of mammalian cell types such as embryonic stem (ES) cells. While it has been generally held that pluripotency is the product of a transcriptional regulatory network that activates and maintains the expression of key stem cell genes, accumulating evidence is pointing to a critical role for epigenetic processes in establishing and safeguarding the pluripotency of ES cells, as well as maintaining the identity of differentiated cell types. In order to better understand the role of epigenetic mechanisms in pluripotency, we have examined the dynamics of chromatin modifications genome-wide in human ES cells (hESCs) undergoing differentiation into a mesendodermal lineage. We found that chromatin modifications at promoters remain largely invariant during differentiation, except at a small number of promoters where a dynamic switch between acetylation and methylation at H3K27 marks the transition between activation and silencing of gene expression, suggesting a hierarchy in cell fate commitment over most differentially expressed genes. We also mapped over 50 000 potential enhancers, and observed much greater dynamics in chromatin modifications, especially H3K4me1 and H3K27ac, which correlate with expression of their potential target genes. Further analysis of these enhancers revealed potentially key transcriptional regulators of pluripotency and a chromatin signature indicative of a poised state that may confer developmental competence in hESCs. Our results provide new evidence supporting the role of chromatin modifications in defining enhancers and pluripotency.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética/fisiologia , Células-Tronco Pluripotentes/metabolismo , Transcrição Gênica/fisiologia , Linhagem Celular , Linhagem da Célula/fisiologia , Cromatina/genética , Cromatina/metabolismo , Células-Tronco Embrionárias/citologia , Elementos Facilitadores Genéticos/fisiologia , Estudo de Associação Genômica Ampla , Humanos , Células-Tronco Pluripotentes/citologia
14.
Cell Stem Cell ; 8(3): 326-34, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21362572

RESUMO

Here, we show that as human embryonic stem cells (ESCs) exit the pluripotent state, NANOG can play a key role in determining lineage outcome. It has previously been reported that BMPs induce differentiation of human ESCs into extraembryonic lineages. Here, we find that FGF2, acting through the MEK-ERK pathway, switches BMP4-induced human ESC differentiation outcome to mesendoderm, characterized by the uniform expression of T (brachyury) and other primitive streak markers. We also find that MEK-ERK signaling prolongs NANOG expression during BMP-induced differentiation, that forced NANOG expression results in FGF-independent BMP4 induction of mesendoderm, and that knockdown of NANOG greatly reduces T induction. Together, our results demonstrate that FGF2 signaling switches the outcome of BMP4-induced differentiation of human ESCs by maintaining NANOG levels through the MEK-ERK pathway.


Assuntos
Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/enzimologia , Endoderma/citologia , Endoderma/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Proteína Homeobox Nanog
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...