Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 84(3): 476-489.e10, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38211589

RESUMEN

Pioneer transcription factors (TFs) regulate cell fate by establishing transcriptionally primed and active states. However, cell fate control requires the coordination of both lineage-specific gene activation and repression of alternative-lineage programs, a process that is poorly understood. Here, we demonstrate that the pioneer TF FOXA coordinates with PRDM1 TF to recruit nucleosome remodeling and deacetylation (NuRD) complexes and Polycomb repressive complexes (PRCs), which establish highly occupied, accessible nucleosome conformation with bivalent epigenetic states, thereby preventing precocious and alternative-lineage gene expression during human endoderm differentiation. Similarly, the pioneer TF OCT4 coordinates with PRDM14 to form bivalent enhancers and repress cell differentiation programs in human pluripotent stem cells, suggesting that this may be a common and critical function of pioneer TFs. We propose that pioneer and PRDM TFs coordinate to safeguard cell fate through epigenetic repression mechanisms.


Asunto(s)
Nucleosomas , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Nucleosomas/genética , Diferenciación Celular/genética , Proteínas del Grupo Polycomb/metabolismo , Epigénesis Genética
2.
J Biol Chem ; 300(3): 105714, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38309502

RESUMEN

Inhibition of protein kinase C (PKC) efficiently promoted the self-renewal of embryonic stem cells (ESCs). However, information about the function of PKC inhibition remains lacking. Here, RNA-sequencing showed that the addition of Go6983 significantly inhibited the expression of de novo methyltransferases (Dnmt3a and Dnmt3b) and their regulator Dnmt3l, resulting in global hypomethylation of DNA in mouse ESCs. Mechanistically, PR domain-containing 14 (Prdm14), a site-specific transcriptional activator, partially contributed to Go6983-mediated repression of Dnmt3 genes. Administration of Go6983 increased Prdm14 expression mainly through the inhibition of PKCδ. High constitutive expression of Prdm14 phenocopied the ability of Go6983 to maintain` mouse ESC stemness in the absence of self-renewal-promoting cytokines. In contrast, the knockdown of Prdm14 eliminated the response to PKC inhibition and substantially impaired the Go6983-induced resistance of mouse ESCs to differentiation. Furthermore, liquid chromatography-mass spectrometry profiling and Western blotting revealed low levels of Suv39h1 and Suv39h2 in Go6983-treated mouse ESCs. Suv39h enzymes are histone methyltransferases that recognize dimethylated and trimethylated histone H3K9 specifically and usually function as transcriptional repressors. Consistently, the inhibition of Suv39h1 by RNA interference or the addition of the selective inhibitor chaetocin increased Prdm14 expression. Moreover, chromatin immunoprecipitation assay showed that Go6983 treatment led to decreased enrichment of dimethylation and trimethylation of H3K9 at the Prdm14 promoter but increased RNA polymerase Ⅱ binding affinity. Together, our results provide novel insights into the pivotal association between PKC inhibition-mediated self-renewal and epigenetic changes, which will help us better understand the regulatory network of stem cell pluripotency.


Asunto(s)
Proteínas de Unión al ADN , Células Madre Embrionarias de Ratones , Proteína Quinasa C , Animales , Ratones , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Indoles/farmacología , Maleimidas/farmacología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/enzimología , Células Madre Embrionarias de Ratones/fisiología , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Inhibidores de Proteínas Quinasas/farmacología
3.
Cell Mol Life Sci ; 81(1): 208, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710919

RESUMEN

Trophoblast stem cells (TSCs) can be chemically converted from embryonic stem cells (ESCs) in vitro. Although several transcription factors (TFs) have been recognized as essential for TSC formation, it remains unclear how differentiation cues link elimination of stemness with the establishment of TSC identity. Here, we show that PRDM14, a critical pluripotent circuitry component, is reduced during the formation of TSCs. The reduction is further shown to be due to the activation of Wnt/ß-catenin signaling. The extinction of PRDM14 results in the erasure of H3K27me3 marks and chromatin opening in the gene loci of TSC TFs, including GATA3 and TFAP2C, which enables their expression and thus the initiation of the TSC formation process. Accordingly, PRDM14 reduction is proposed here as a critical event that couples elimination of stemness with the initiation of TSC formation. The present study provides novel insights into how induction signals initiate TSC formation.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN , Factores de Transcripción , Trofoblastos , Vía de Señalización Wnt , Trofoblastos/metabolismo , Trofoblastos/citología , Animales , Ratones , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Factor de Transcripción GATA3/metabolismo , Factor de Transcripción GATA3/genética , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Células Madre/metabolismo , Células Madre/citología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Histonas/metabolismo , Histonas/genética
4.
BMC Genomics ; 25(1): 344, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38580899

RESUMEN

BACKGROUND: Genome-wide DNA demethylation occurs in mammalian primordial germ cells (PGCs) as part of the epigenetic reprogramming important for gametogenesis and resetting the epigenetic information for totipotency. Dppa3 (also known as Stella or Pgc7) is highly expressed in mouse PGCs and oocytes and encodes a factor essential for female fertility. It prevents excessive DNA methylation in oocytes and ensures proper gene expression in preimplantation embryos: however, its role in PGCs is largely unexplored. In the present study, we investigated whether or not DPPA3 has an impact on CG methylation/demethylation in mouse PGCs. RESULTS: We show that DPPA3 plays a role in genome-wide demethylation in PGCs even before sex differentiation. Dppa3 knockout female PGCs show aberrant hypermethylation, most predominantly at H3K9me3-marked retrotransposons, which persists up to the fully-grown oocyte stage. DPPA3 works downstream of PRDM14, a master regulator of epigenetic reprogramming in embryonic stem cells and PGCs, and independently of TET1, an enzyme that hydroxylates 5-methylcytosine. CONCLUSIONS: The results suggest that DPPA3 facilitates DNA demethylation through a replication-coupled passive mechanism in PGCs. Our study identifies DPPA3 as a novel epigenetic reprogramming factor in mouse PGCs.


Asunto(s)
Proteínas Cromosómicas no Histona , Desmetilación del ADN , Epigénesis Genética , Animales , Femenino , Ratones , Proteínas Cromosómicas no Histona/metabolismo , Metilación de ADN , Genoma , Células Germinativas/metabolismo , Mamíferos/genética
5.
BMC Cancer ; 24(1): 685, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38840106

RESUMEN

BACKGROUND: Gastric cancer is one of the most common tumors worldwide, and most patients are deprived of treatment options when diagnosed at advanced stages. PRDM14 has carcinogenic potential in breast and non-small cell lung cancer. however, its role in gastric cancer has not been elucidated. METHODS: We aimed to elucidate the expression of PRDM14 using pan-cancer analysis. We monitored the expression of PRDM14 in cells and patients using quantitative polymerase chain reaction, western blotting, and immunohistochemistry. We observed that cell phenotypes and regulatory genes were influenced by PRDM14 by silencing PRDM14. We evaluated and validated the value of the PRDM14-derived prognostic model. Finally, we predicted the relationship between PRDM14 and small-molecule drug responses using the Connectivity Map and The Genomics of Drug Sensitivity in Cancer databases. RESULTS: PRDM14 was significantly overexpressed in gastric cancer, which identified in cell lines and patients' tissues. Silencing the expression of PRDM14 resulted in apoptosis promotion, cell cycle arrest, and inhibition of the growth and migration of GC cells. Functional analysis revealed that PRDM14 acts in epigenetic regulation and modulates multiple DNA methyltransferases or transcription factors. The PRDM14-derived differentially expressed gene prognostic model was validated to reliably predict the patient prognosis. Nomograms (age, sex, and PRDM14-risk score) were used to quantify the probability of survival. PRDM14 was positively correlated with sensitivity to small-molecule drugs such as TPCA-1, PF-56,227, mirin, and linsitinib. CONCLUSIONS: Collectively, our findings suggest that PRDM14 is a positive regulator of gastric cancer progression. Therefore, it may be a potential therapeutic target for gastric cancer.


Asunto(s)
Proteínas de Unión al ADN , Regulación Neoplásica de la Expresión Génica , Neoplasias Gástricas , Factores de Transcripción , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Pronóstico , Línea Celular Tumoral , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Femenino , Masculino , Nomogramas , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Epigénesis Genética
6.
Development ; 147(4)2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-32001439

RESUMEN

Primordial germ cells (PGCs), the founder cells of the germline, are specified in pre-gastrulating embryos in mammals, and subsequently migrate towards gonads to mature into functional gametes. Here, we investigated PGC development in rats, by genetically modifying Prdm14, a unique marker and an essential PGC transcriptional regulator. We trace PGC development in rats, for the first time, from specification until the sex determination stage in fetal gonads using Prdm14 H2BVenus knock-in rats. We uncover that the crucial role of Prdm14 in PGC specification is conserved between rat and mice, by analyzing Prdm14-deficient rat embryos. Notably, loss of Prdm14 completely abrogates the PGC program, as demonstrated by failure of the maintenance and/or activation of germ cell markers and pluripotency genes. Finally, we profile the transcriptome of the post-implantation epiblast and all PGC stages in rat to reveal enrichment of distinct gene sets at each transition point, thereby providing an accurate transcriptional timeline for rat PGC development. Thus, the novel genetically modified rats and data sets obtained in this study will advance our knowledge on conserved versus species-specific features for germline development in mammals.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Germinativas/citología , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética , Animales , Cruzamientos Genéticos , Proteínas de Unión al ADN/fisiología , Femenino , Gástrula/fisiología , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Masculino , Ratones , Proteínas de Unión al ARN/fisiología , Ratas , Procesos de Determinación del Sexo , Factores de Transcripción/fisiología , Transcripción Genética
7.
J Biol Chem ; 297(4): 101217, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34555410

RESUMEN

Primordial germ cells (PGCs) are common ancestors of all germline cells. However, mechanistic understanding of how PGC specification occurs is limited. Here, we identified transcription factor CP2-like 1 (Tfcp2l1), an important pluripotency factor, as a pivotal factor for PGC-like cell (PGCLC) specification. High-throughput sequencing and quantitative real-time PCR analysis showed that Tfcp2l1 expression is gradually increased during mouse and human epiblast differentiation into PGCLCs in vivo and in vitro. Consequently, overexpression of Tfcp2l1 can enhance the specification efficiency even without inductive cytokines in mouse epiblast-like cells derived from embryonic stem cells, while knockdown of Tfcp2l1 significantly inhibits PGCLC generation. Mechanistic studies revealed that Tfcp2l1 exerts its function partially through the direct induction of PR domain zinc finger protein 14, a key PGC marker, as downregulation of the PR domain zinc finger protein 14 transcript can impair the ability of Tfcp2l1 to direct PGCLC commitment. Importantly, we finally demonstrated that the crucial role of the human homolog Tfcp2l1 in promoting PGCLC specification is conserved in human pluripotent stem cells. Together, our data uncover a novel function of Tfcp2l1 in PGCLC fate determination and facilitate a better understanding of germ cell development.


Asunto(s)
Células Madre Pluripotentes/metabolismo , Proteínas Represoras/metabolismo , Animales , Línea Celular , Células Germinativas , Humanos , Ratones , Dominios Proteicos , Proteínas Represoras/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
8.
Trends Genet ; 35(7): 489-500, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31130394

RESUMEN

Despite advances in chemotherapies that improve cancer survival, most patients who relapse succumb to the disease due to the presence of cancer stem cells (CSCs), which are highly chemoresistant. The pluripotency factor PR domain 14 (PRDM14) has a key role in initiating many types of cancer. Normally, PRDM14 uses epigenetic mechanisms to establish and maintain the pluripotency of embryonic cells, and its role in cancer is similar. This important link between cancer and induced pluripotency is a key revelation for how CSCs may form: pluripotency genes, such as PRDM14, can expand stem-like cells as they promote ongoing DNA damage. PRDM14 and its protein-binding partners, the ETO/CBFA2T family, are ideal candidates for eliminating CSCs from relevant cancers, preventing relapse and improving long-term survival.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Neoplasias/genética , Células Madre Pluripotentes/patología , Proteínas de Unión al ARN/fisiología , Factores de Transcripción/fisiología , Daño del ADN , Epigénesis Genética , Inestabilidad Genómica , Humanos , Neoplasias/patología
9.
J Cell Sci ; 133(15)2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32661086

RESUMEN

The pluripotency-associated transcriptional network is regulated by a core circuitry of transcription factors. The PR domain-containing protein PRDM14 maintains pluripotency by activating and repressing transcription in a target gene-dependent manner. However, the mechanisms underlying dichotomic switching of PRDM14-mediated transcriptional control remain elusive. Here, we identified C-terminal binding protein 1 and 2 (CtBP1 and CtBP2; generically referred to as CtBP1/2) as components of the PRDM14-mediated repressive complex. CtBP1/2 binding to PRDM14 depends on CBFA2T2, a core component of the PRDM14 complex. The loss of Ctbp1/2 impaired the PRDM14-mediated transcriptional repression required for pluripotency maintenance and transition from primed to naïve pluripotency. Furthermore, CtBP1/2 interacted with the PRC2 complexes, and the loss of Ctbp1/2 impaired Polycomb repressive complex 2 (PRC2) and H3K27me3 enrichment at target genes after Prdm14 induction. These results provide evidence that the target gene-dependent transcriptional activity of PRDM14 is regulated by partner switching to ensure the transition from primed to naïve pluripotency.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Proteínas de Unión al ADN , Complejo Represivo Polycomb 2 , Oxidorreductasas de Alcohol/genética , Proteínas Co-Represoras , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Complejo Represivo Polycomb 2/metabolismo , Proteínas de Unión al ARN , Factores de Transcripción
10.
Development ; 146(2)2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30630825

RESUMEN

Gene regulatory networks underlying cellular pluripotency are controlled by a core circuitry of transcription factors in mammals, including POU5F1. However, the evolutionary origin and transformation of pluripotency-related transcriptional networks have not been elucidated in deuterostomes. PR domain-containing protein 14 (PRDM14) is specifically expressed in pluripotent cells and germ cells, and is required for establishing embryonic stem cells (ESCs) and primordial germ cells in mice. Here, we compared the functions and expression patterns of PRDM14 orthologues within deuterostomes. Amphioxus PRDM14 and zebrafish PRDM14, but not sea urchin PRDM14, compensated for mouse PRDM14 function in maintaining mouse ESC pluripotency. Interestingly, sea urchin PRDM14 together with sea urchin CBFA2T, an essential partner of PRDM14 in mouse ESCs, complemented the self-renewal defect in mouse Prdm14 KO ESCs. Contrary to the Prdm14 expression pattern in mouse embryos, Prdm14 was expressed in motor neurons of amphioxus embryos, as observed in zebrafish embryos. Thus, Prdm14 expression in motor neurons was conserved in non-tetrapod deuterostomes and the co-option of the PRDM14-CBFA2T complex from motor neurons into pluripotent cells may have maintained the transcriptional network for pluripotency during vertebrate evolution.This article has an associated 'The people behind the papers' interview.


Asunto(s)
Evolución Biológica , Neuronas Motoras/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Vertebrados/metabolismo , Secuencia de Aminoácidos , Animales , Biomarcadores/metabolismo , Desmetilación del ADN , Metilación de ADN , Proteínas de Unión al ADN , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Anfioxos/embriología , Anfioxos/metabolismo , Ratones , Ratones Noqueados , Filogenia , Unión Proteica , Dominios Proteicos , Proteínas de Unión al ARN , Proteínas Represoras/química , Erizos de Mar/embriología , Erizos de Mar/metabolismo , Homología de Secuencia de Ácido Nucleico , Sintenía/genética , Vertebrados/embriología , Pez Cebra/embriología , Pez Cebra/metabolismo
11.
Proc Jpn Acad Ser B Phys Biol Sci ; 98(7): 325-335, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35908955

RESUMEN

PRDI-BF1 and RIZ (PR) domain zinc finger protein 14 (PRDM14), first reported in 2007 to be overexpressed in breast cancer, plays an important role in breast cancer proliferation. Subsequent studies reported that PRDM14 is expressed in embryonic stem cells, primordial germ cells, and various cancers. PRDM14 was reported to confer stemness properties to cancer cells. These properties induce cancer initiation, cancer progression, therapeutic resistance, distant metastasis, and recurrence in refractory tumors. Therefore, PRDM14 may be an ideal therapeutic target for various types of tumors. Silencing PRDM14 expression using PRDM14-specific siRNA delivered through an innovative intravenous drug delivery system reduced the size of inoculated tumors, incidence of distant metastases, and increased overall survival in nude mice without causing adverse effects. Therapeutic siRNA targeting PRDM14 is now being evaluated in a human phase I clinical trial for patients with refractory breast cancer, including triple-negative breast cancer.


Asunto(s)
Neoplasias de la Mama , Proteínas de Unión al ADN , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , ARN Interferente Pequeño , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Dedos de Zinc
12.
Development ; 145(20)2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30185410

RESUMEN

The precise control of gene expression by transcription factor networks is crucial to organismal development. The predominant approach for mapping transcription factor-chromatin interactions has been chromatin immunoprecipitation (ChIP). However, ChIP requires a large number of homogeneous cells and antisera with high specificity. A second approach, DamID, has the drawback that high levels of Dam methylase are toxic. Here, we modify our targeted DamID approach (TaDa) to enable cell type-specific expression in mammalian systems, generating an inducible system (mammalian TaDa or MaTaDa) to identify genome-wide protein/DNA interactions in 100 to 1000 times fewer cells than ChIP-based approaches. We mapped the binding sites of two key pluripotency factors, OCT4 and PRDM14, in mouse embryonic stem cells, epiblast-like cells and primordial germ cell-like cells (PGCLCs). PGCLCs are an important system for elucidating primordial germ cell development in mice. We monitored PRDM14 binding during the specification of PGCLCs, identifying direct targets of PRDM14 that are key to understanding its crucial role in PGCLC development. We show that MaTaDa is a sensitive and accurate method for assessing cell type-specific transcription factor binding in limited numbers of cells.


Asunto(s)
Metilación de ADN/genética , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Cromatina/metabolismo , Proteínas de Unión al ADN , Genoma , Células Germinativas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Unión Proteica , Proteínas de Unión al ARN
13.
Dev Biol ; 455(1): 32-41, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31271752

RESUMEN

The differentiation of primordial germ cells (PGCs) is a fundamental step in development. PR domain-containing protein 14 (PRDM14) and B lymphocyte-induced maturation protein 1 (BLIMP1) play pivotal roles in mouse PGC specification. In the present study, we assessed the roles of chicken orthologs of PRDM14 and BLIMP1 in PGC development. PRDM14 and BLIMP1 were expressed in blastodermal cells and PGCs. The in vivo knockdown of PRDM14 or BLIMP1 by introducing a replication-competent retroviral vector expressing shRNAs to the blastodermal stage of embryos reduced the number of SSEA-1 or chicken vasa homologue-positive PGCs on day 5.5-6.5. Since the inhibition of Activin receptor-like kinase 4/5/7 in cultured PGCs reduced the expression of PRDM14, BLIMP1, and NANOG, and that of MEK inhibited PRDM14 expression, the expression of these genes seems to be controlled by Activin A and FGF2 signaling. Overall, PRDM14, BLIMP1, and NANOG seem to be involved in the self-renewal of PGCs in cultured PGCs and embryos.


Asunto(s)
Proteínas Aviares/genética , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Animales , Proteínas Aviares/metabolismo , Blastodermo/citología , Blastodermo/metabolismo , Autorrenovación de las Células/genética , Células Cultivadas , Embrión de Pollo , Pollos , Células Germinativas/citología , Antígeno Lewis X/genética , Antígeno Lewis X/metabolismo , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Interferencia de ARN
14.
Proc Natl Acad Sci U S A ; 113(44): 12478-12483, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27738243

RESUMEN

Developmental signaling molecules are used for cell fate determination, and understanding how their combinatorial effects produce the variety of cell types in multicellular organisms is a key problem in biology. Here, we demonstrate that the combination of leukemia inhibitory factor (LIF), bone morphogenetic protein 4 (BMP4), lysophosphatidic acid (LPA), and ascorbic acid (AA) efficiently converts mouse primed pluripotent stem cells (PSCs) into naive PSCs. Signaling by the lipid LPA through its receptor LPAR1 and downstream effector Rho-associated protein kinase (ROCK) cooperated with LIF signaling to promote this conversion. BMP4, which also stimulates conversion to naive pluripotency, bypassed the need for exogenous LPA by increasing the activity of the extracellular LPA-producing enzyme autotaxin (ATX). We found that LIF and LPA-LPAR1 signaling affect the abundance of signal transducer and activator of transcription 3 (STAT3), which induces a previously unappreciated Kruppel-like factor (KLF)2-KLF4-PR domain 14 (PRDM14) transcription factor circuit key to establish naive pluripotency. AA also affects this transcription factor circuit by controlling PRDM14 expression. Thus, our study reveals that ATX-mediated autocrine lipid signaling promotes naive pluripotency by intersecting with LIF and BMP4 signaling.


Asunto(s)
Proteína Morfogenética Ósea 4/farmacología , Factor Inhibidor de Leucemia/farmacología , Lisofosfolípidos/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , Células Madre Pluripotentes/efectos de los fármacos , Factores de Transcripción/metabolismo , Animales , Ácido Ascórbico/farmacología , Línea Celular , Reprogramación Celular/efectos de los fármacos , Reprogramación Celular/genética , Sinergismo Farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Factor 4 Similar a Kruppel , Ratones Endogámicos C57BL , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/genética , Vitaminas/farmacología
15.
Trends Biochem Sci ; 39(6): 289-98, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24811060

RESUMEN

PRDM14 belongs to the PR domain-containing (PRDM) transcriptional regulators. Among the PRDM family members, PRDM14 shows specific expression in preimplantation embryos, primordial germ cells (PGCs), and embryonic stem cells (ESCs) in vitro, and accordingly plays a key role in the regulation of their pluripotency and epigenetic reprogramming, most notably, genome-wide DNA demethylation. The function of PRDM14 appears to be conserved between mice and humans, but it shows several characteristic differences between the two species. A precise understanding of the function of PRDM14 in mice and humans would shed new light on the regulation of pluripotency and the epigenome in these two species, providing a foundation for better control of stem cell fates in a broader context.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN , Humanos , Ratones , Proteínas de Unión al ARN
16.
Cancer Sci ; 109(2): 373-383, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29178343

RESUMEN

PRDM14 is overexpressed in various cancers and can regulate cancer phenotype under certain conditions. Inhibiting PRDM14 expression in breast and pancreatic cancers has been reported to reduce cancer stem-like phenotypes, which are associated with aggressive tumor properties. Therefore, PRDM14 is considered a promising target for cancer therapy. To develop a pharmaceutical treatment, the mechanism and interacting partners of PRDM14 need to be clarified. Here, we identified the proteins interacting with PRDM14 in triple-negative breast cancer (TNBC) cells, which do not express the three most common types of receptor (estrogen receptors, progesterone receptors, and HER2). We obtained 13 candidates that were pulled down with PRDM14 in TNBC HCC1937 cells and identified them by mass spectrometry. Two candidates-glucose-regulated protein 78 (GRP78) and heat shock protein 90-α (HSP90α)-were confirmed in immunoprecipitation assay in two TNBC cell lines (HCC1937 and MDA-MB231). Surface plasmon resonance analysis using GST-PRDM14 showed that these two proteins directly interacted with PRDM14 and that the interactions required the C-terminal region of PRDM14, which includes zinc finger motifs. We also confirmed the interactions in living cells by NanoLuc luciferase-based bioluminescence resonance energy transfer (NanoBRET) assay. Moreover, HSP90 inhibitors (17DMAG and HSP990) significantly decreased breast cancer stem-like CD24-  CD44+ and side population (SP) cells in HCC1937 cells, but not in PRDM14 knockdown HCC1937 cells. The combination of the GRP78 inhibitor HA15 and PRDM14 knockdown significantly decreased cell proliferation and SP cell number in HCC1937 cells. These results suggest that HSP90α and GRP78 interact with PRDM14 and participate in cancer regulation.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Represoras/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN , Chaperón BiP del Retículo Endoplásmico , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Espectrometría de Masas , Dominios Proteicos , Mapeo de Interacción de Proteínas , Proteínas de Unión al ARN , Proteínas Represoras/química , Resonancia por Plasmón de Superficie , Factores de Transcripción
17.
Stem Cells ; 34(2): 322-33, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26523946

RESUMEN

Identification of a gene set capable of driving rapid and proper reprogramming to induced pluripotent stem cells (iPSCs) is an important issue. Here we show that the efficiency and kinetics of iPSC reprogramming are dramatically improved by the combined expression of Jarid2 and genes encoding its associated proteins. We demonstrate that forced expression of JARID2 promotes iPSC reprogramming by suppressing the expression of Arf, a known reprogramming barrier, and that the N-terminal half of JARID2 is sufficient for such promotion. Moreover, JARID2 accelerated silencing of the retroviral Klf4 transgene and demethylation of the Nanog promoter, underpinning the potentiating activity of JARID2 in iPSC reprogramming. We further show that JARID2 physically interacts with ESRRB, SALL4A, and PRDM14, and that these JARID2-associated proteins synergistically and robustly facilitate iPSC reprogramming in a JARID2-dependent manner. Our findings provide an insight into the important roles of JARID2 during reprogramming and suggest that the JARID2-associated protein network contributes to overcoming reprogramming barriers.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Proteínas de Unión al ADN , Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Complejo Represivo Polycomb 2 , Receptores de Estrógenos , Factores de Transcripción , Animales , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Factor 4 Similar a Kruppel , Ratones , Complejo Represivo Polycomb 2/biosíntesis , Complejo Represivo Polycomb 2/genética , Proteínas de Unión al ARN , Receptores de Estrógenos/biosíntesis , Receptores de Estrógenos/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
18.
Dev Dyn ; 245(7): 807-15, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27012147

RESUMEN

BACKGROUND: Pluripotent embryonic stem cells (ESCs) offer great potential for regenerative medicine. However, efficient in vitro generation of specific desired cell types is still a challenge. We previously established that Smad2/3 signaling, essential for endoderm formation, regulates target gene expression by counteracting epigenetic repression mediated by Polycomb Repressive Complex 2 (PRC2). Although this mechanism has been demonstrated during differentiation and reprogramming, little is known of its role in pluripotent cells. RESULTS: Chromatin immunoprecipitation-deep sequencing of undifferentiated mouse ESCs inhibited for Smad2/3 signaling identified Prdm14, important for protecting pluripotency, as a target gene. Although Prdm14 accumulates the normally repressive PRC2 deposited histone modification H3K27me3 under these conditions, surprisingly, expression increases. Analysis indicates that increased H3K27me3 leads to increased binding of PRC2 accessory component Jarid2 and recruitment of RNA polymerase II. Similar increases were found at the Nodal endoderm target gene Eomes but it remained unexpressed in pluripotent cells as normal. Upon differentiation, however, Eomes expression was significantly higher than in cells that had not been inhibited for signaling before differentiation. In addition, endoderm formation was markedly increased. CONCLUSIONS: Blocking Smad2/3 signaling in pluripotent stem cells results in epigenetic changes that enhance the capacity for endoderm differentiation. Developmental Dynamics 245:807-815, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Endodermo/citología , Endodermo/metabolismo , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN , Ratones , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Unión Proteica , Proteínas de Unión al ARN , Proteína Smad2/genética , Proteína smad3/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Biochem Biophys Res Commun ; 466(1): 138-45, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26325469

RESUMEN

Pluripotency and self-renewal of mouse embryonic stem cells (ESCs) depend on a network of transcription factors maintained by exogenous leukaemia inhibitory factor (LIF). PR-domain containing transcriptional regulator 14 (PRDM14), is essential for maintenance of ESC self-renewal when the cells are cultured in serum plus LIF, but not in 2i medium plus LIF. Here, we show that pluripotency of ESCs is maintained by enforced expression of PRDM14 at a high level, as observed in ESCs in 2i plus LIF and developing primordial germ cells in the absence of LIF. Constitutive expression of PRDM14 represses de novo DNA methylation in pluripotency-associated genes, resulting in the maintenance of gene expression after withdrawal of LIF, while also repressing the upregulation of differentiation markers. Further, knockdown of Tet1/Tet2 and administration of base excision repair (BER) pathway inhibitors impairs the PRDM14-induced resistance of ESCs to differentiation. We conclude that, in the absence of LIF, PRDM14 governs the retention of pluripotency-associated genes through the regulation of TET functions in the BER-mediated active demethylation pathway, while acting to exert TET-independent transcriptional repressive activity of several differentiation markers.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/citología , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/genética , Animales , Diferenciación Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Dioxigenasas , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Factor Inhibidor de Leucemia/metabolismo , Ratones , Proteínas Proto-Oncogénicas/genética , Proteínas de Unión al ARN , Factores de Transcripción/metabolismo , Regulación hacia Arriba
20.
RNA Biol ; 11(7): 798-807, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25137047

RESUMEN

X-chromosome inactivation (XCI) in female mammals is a dramatic example of epigenetic gene regulation, which entails the silencing of an entire chromosome through a wide range of mechanisms involving noncoding RNAs, chromatin-modifications, and DNA-methylation. While XCI is associated with the differentiated cell state, it is reversed by X-chromosome reactivation (XCR) ex vivo in pluripotent stem cells and in vivo in the early mouse embryo and the germline. Critical in the regulation of XCI vs. XCR is the X-inactivation center, a multigene locus on the X-chromosome harboring several long noncoding RNA genes including, most prominently, Xist and Tsix. These genes, which sit at the top of the XCI hierarchy, are by themselves controlled by pluripotency factors, coupling XCR with the naïve pluripotent stem cell state. In this point-of-view article we review the latest findings regarding this intricate relationship between cell differentiation state and epigenetic control of the X-chromosome. In particular, we discuss the emerging picture of complex multifactorial regulatory mechanisms, ensuring both a fine-tuned and robust X-reactivation process.


Asunto(s)
Células Madre Pluripotentes/metabolismo , ARN Largo no Codificante/metabolismo , Factores de Transcripción/metabolismo , Inactivación del Cromosoma X , Animales , Diferenciación Celular , Femenino , Humanos , Ratones , Cromosoma X/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA