Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Biosci ; 13(1): 88, 2023 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-37194020

RESUMEN

BACKGROUND: Extended pluripotent stem cells (EPSCs) can contribute to both embryonic and trophectoderm-derived extraembryonic tissues. Therefore, EPSCs have great application significance for both research and industry. However, generating EPSCs from human somatic cells remains inefficient and cumbersome. RESULTS: In this study, we established a novel and robust EPSCs culture medium OCM175 with defined and optimized ingredients. Our OCM175 medium contains optimized concentration of L-selenium-methylcysteine as a source of selenium and ROCK inhibitors to maintain the single cell passaging ability of pluripotent stem cells. We also used Matrigel or the combination of laminin 511 and laminin 521(1:1) to bypass the requirement of feeder cells. With OCM175 medium, we successfully converted integration-free iPSCs from easily available human Urine-Derived Cells (hUC-iPSCs) into EPSCs (O-IPSCs). We showed that our O-IPSCs have the ability to form both intra- and extra- embryonic chimerism, and could contribute to the trophoblast ectoderm lineage and three germ layer cell lineages. CONCLUSIONS: In conclusion, our novel OCM175 culture medium has defined, optimized ingredients, which enables efficient generation of EPSCs in a feeder free manner. With the robust chimeric and differentiation potential, we believe that this system provides a solid basis to improve the application of EPSCs in regenerative medicine.

3.
Nat Commun ; 12(1): 4090, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34215745

RESUMEN

The transition from pluripotent to somatic states marks a critical event in mammalian development, but remains largely unresolved. Here we report the identification of SS18 as a regulator for pluripotent to somatic transition or PST by CRISPR-based whole genome screens. Mechanistically, SS18 forms microscopic condensates in nuclei through a C-terminal intrinsically disordered region (IDR) rich in tyrosine, which, once mutated, no longer form condensates nor rescue SS18-/- defect in PST. Yet, the IDR alone is not sufficient to rescue the defect even though it can form condensates indistinguishable from the wild type protein. We further show that its N-terminal 70aa is required for PST by interacting with the Brg/Brahma-associated factor (BAF) complex, and remains functional even swapped onto unrelated IDRs or even an artificial 24 tyrosine polypeptide. Finally, we show that SS18 mediates BAF assembly through phase separation to regulate PST. These studies suggest that SS18 plays a role in the pluripotent to somatic interface and undergoes liquid-liquid phase separation through a unique tyrosine-based mechanism.


Asunto(s)
Transición de Fase , Células Madre Pluripotentes/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Animales , Núcleo Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Femenino , Células HEK293 , Humanos , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Tirosina
4.
Nat Cell Biol ; 22(6): 651-662, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32393886

RESUMEN

BMP4 regulates a plethora of developmental processes, including the dorsal-ventral axis and neural patterning. Here, we report that BMP4 reconfigures the nuclear architecture during the primed-to-naive transition (PNT). We first established a BMP4-driven PNT and show that BMP4 orchestrates the chromatin accessibility dynamics during PNT. Among the loci opened early by BMP4, we identified Zbtb7a and Zbtb7b (Zbtb7a/b) as targets that drive PNT. ZBTB7A/B in turn facilitate the opening of naive pluripotent chromatin loci and the activation of nearby genes. Mechanistically, ZBTB7A not only binds to chromatin loci near to the genes that are activated, but also strategically occupies those that are silenced, consistent with a role of BMP4 in both activating and suppressing gene expression during PNT at the chromatin level. Our results reveal a previously unknown function of BMP4 in regulating nuclear architecture and link its targets ZBTB7A/B to chromatin remodelling and pluripotent fate control.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/citología , Estratos Germinativos/citología , Células Madre Pluripotentes/citología , Factores de Transcripción/metabolismo , Animales , Blastocisto/citología , Blastocisto/metabolismo , Proteína Morfogenética Ósea 4/genética , Diferenciación Celular , Células Cultivadas , Cromatina/genética , Proteínas de Unión al ADN/genética , Células Madre Embrionarias/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Células Madre Pluripotentes/metabolismo , Transducción de Señal , Factores de Transcripción/genética
5.
Cell Rep ; 27(12): 3473-3485.e5, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31216469

RESUMEN

Reprogramming somatic cells to pluripotency by Oct4, Sox2, Klf4, and Myc represent a paradigm for cell fate determination. Here, we report a combination of Jdp2, Jhdm1b, Mkk6, Glis1, Nanog, Essrb, and Sall4 (7F) that reprogram mouse embryonic fibroblasts or MEFs to chimera competent induced pluripotent stem cells (iPSCs) efficiently. RNA sequencing (RNA-seq) and ATAC-seq reveal distinct mechanisms for 7F induction of pluripotency. Dropout experiments further reveal a highly cooperative process among 7F to dynamically close and open chromatin loci that encode a network of transcription factors to mediate reprogramming. These results establish an alternative paradigm for reprogramming that may be useful for analyzing cell fate control.


Asunto(s)
Reprogramación Celular/genética , Proteínas de Unión al ADN/metabolismo , Proteínas F-Box/metabolismo , Células Madre Pluripotentes Inducidas/citología , Histona Demetilasas con Dominio de Jumonji/metabolismo , MAP Quinasa Quinasa 6/metabolismo , Proteína Homeótica Nanog/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Quimera/genética , Proteínas de Unión al ADN/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Eucromatina/genética , Eucromatina/metabolismo , Proteínas F-Box/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Heterocromatina/genética , Heterocromatina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Factor 4 Similar a Kruppel , MAP Quinasa Quinasa 6/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Homeótica Nanog/genética , RNA-Seq , Proteínas Represoras/genética , Factores de Transcripción/genética
6.
Mol Cell ; 73(4): 815-829.e7, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30772174

RESUMEN

Somatic cells can be reprogrammed into induced pluripotent stem cells (iPSCs), which is a highly heterogeneous process. Here we report the cell fate continuum during somatic cell reprogramming at single-cell resolution. We first develop SOT to analyze cell fate continuum from Oct4/Sox2/Klf4- or OSK-mediated reprogramming and show that cells bifurcate into two categories, reprogramming potential (RP) or non-reprogramming (NR). We further show that Klf4 contributes to Cd34+/Fxyd5+/Psca+ keratinocyte-like NR fate and that IFN-γ impedes the final transition to chimera-competent pluripotency along the RP cells. We analyze more than 150,000 single cells from both OSK and chemical reprograming and identify additional NR/RP bifurcation points. Our work reveals a generic bifurcation model for cell fate decisions during somatic cell reprogramming that may be applicable to other systems and inspire further improvements for reprogramming.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Técnicas de Reprogramación Celular , Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Embrionarias de Ratones/fisiología , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Interferón gamma/genética , Interferón gamma/metabolismo , Factor 4 Similar a Kruppel , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Células Madre Embrionarias de Ratones/metabolismo , Fenotipo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Nat Commun ; 9(1): 4649, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30405129

RESUMEN

Human pluripotent stem cells (hPSCs) exhibit very limited contribution to interspecies chimeras. One explanation is that the conventional hPSCs are in a primed state and so unable  to form chimeras in pre-implantation embryos. Here, we show that the conventional hPSCs undergo rapid apoptosis when injected into mouse pre-implantation embryos. While, forced-expression of BMI1, a polycomb factor in hPSCs overcomes the apoptosis and enables hPSCs to integrate into mouse pre-implantation embryos and subsequently contribute to chimeras with both embryonic and extra-embryonic tissues. In addition, BMI1 also enables hPSCs to integrate into pre-implantation embryos of other species, such as rabbit and pig. Notably, BMI1 high expression and anti-apoptosis are also indicators for naïve hPSCs to form chimera in mouse embryos. Together, our findings reveal that the apoptosis is an initial barrier in interspecies chimerism using hPSCs and provide a rational to improve it.


Asunto(s)
Quimerismo , Células Madre Pluripotentes/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Animales , Apoptosis , Blastocisto/citología , Blastocisto/metabolismo , Linaje de la Célula , Membranas Extraembrionarias/metabolismo , Humanos , Ratones Endogámicos ICR , Células Madre Pluripotentes/citología , Conejos , Especificidad de la Especie , Porcinos
9.
J Biol Chem ; 292(46): 19122-19132, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-28935668

RESUMEN

We report here an approach to redirecting somatic cell fate under chemically defined conditions without transcription factors. We start by converting mouse embryonic fibroblasts to epithelial-like cells with chemicals and growth factors. Subsequent cell fate mapping reveals a robust induction of SOX17 in the resulting epithelial-like cells that can be further reprogrammed to endodermal progenitor cells. Interestingly, these cells can self-renew in vitro and further differentiate into albumin-producing hepatocytes that can rescue mice from acute liver injury. Our results demonstrate a rational approach to convert mouse embryonic fibroblasts to hepatocytes and suggest that this mechanism-driven approach may be generalized for other cells.


Asunto(s)
Reprogramación Celular/efectos de los fármacos , Endodermo/citología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Células Madre/citología , Animales , Diferenciación Celular , Autorrenovación de las Células , Células Cultivadas , Femenino , Proteínas HMGB/análisis , Hepatocitos/citología , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción SOXF/análisis
10.
Cell Res ; 21(6): 884-94, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21445094

RESUMEN

The ectopic expression of several transcription factors can restore embryonic cell fate to cultured somatic cells and generate induced pluripotent stem cells (iPSCs), revealing a previously unknown pathway to pluripotency. However, this technology is currently limited by low efficiency, slow kinetics and multi-factorial requirement. Here we show that reprogramming can be improved and dramatically accelerated by optimizing culture conditions. First, we developed an optimized defined medium, iCD1, which allows Oct4/Sox2/Klf4 (OSK)-mediated reprogramming to achieve ultra-high efficiency (~10% at day 8). We also found that this optimized condition renders both Sox2 and Klf4 dispensable, although the elimination of these two factors leads to lower efficiency and slower kinetics. Our studies define a shortened route, both in timing and factor requirement, toward pluripotency. This new paradigm not only provides a rationale to further improve iPSC generation but also simplifies the conceptual understanding of reprogramming by defined factors.


Asunto(s)
Desdiferenciación Celular/genética , Medios de Cultivo/química , Células Madre Pluripotentes Inducidas/citología , Animales , Técnicas de Cultivo de Célula , Proliferación Celular , Técnicas de Cocultivo , Medios de Cultivo/metabolismo , Fibroblastos/metabolismo , Vectores Genéticos , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Cariotipificación , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Endogámicos ICR , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Retroviridae/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Tiempo , Transfección , Quimera por Trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA