Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Dev Biol ; 478: 222-235, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34246625

RESUMO

Peroxisome proliferator-activated receptor (PPAR) γ1, a nuclear receptor, is abundant in the murine placenta during the late stage of pregnancy (E15-E16), although its functional roles remain unclear. PPARγ1 is encoded by two splicing isoforms, namely Pparγ1canonical and Pparγ1sv, and its embryonic loss leads to early (E10) embryonic lethality. Thus, we generated knockout (KO) mice that carried only one of the isoforms to obtain a milder phenotype. Pparγ1sv-KO mice were viable and fertile, whereas Pparγ1canonical-KO mice failed to recover around the weaning age. Pparγ1canonical-KO embryos developed normally up to 15.5 dpc, followed by growth delays after that. The junctional zone of Pparγ1canonical-KO placentas severely infiltrated the labyrinth, and maternal blood sinuses were dilated. In the wild-type, PPARγ1 was highly expressed in sinusoidal trophoblast giant cells (S-TGCs), peaking at 15.5 dpc. Pparγ1canonical-KO abolished PPARγ1 expression in S-TGCs. Notably, the S-TGCs had unusually enlarged nuclei and often occupied maternal vascular spaces, disturbing the organization of the fine labyrinth structure. Gene expression analyses of Pparγ1canonical-KO placentas indicated enhanced S-phase cell cycle signatures. EdU-positive S-TGCs in Pparγ1canonical-KO placentas were greater in number than those in wild-type placentas, suggesting that the cells continued to endoreplicate in the mutant placentas. These results indicate that PPARγ1, a known cell cycle arrest mediator, is involved in the transition of TGCs undergoing endocycling to the terminal differentiation stage in the placentas. Therefore, PPARγ1 deficiency, induced through genetic manipulation, leads to placental insufficiency.


Assuntos
Ciclo Celular , Desenvolvimento Embrionário , Endorreduplicação , PPAR gama/genética , PPAR gama/metabolismo , Placenta/metabolismo , Trofoblastos/citologia , Animais , Diferenciação Celular , Feminino , Retardo do Crescimento Fetal , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placenta/anormalidades , Placenta/citologia , Insuficiência Placentária/etiologia , Gravidez , Transcrição Gênica , Trofoblastos/metabolismo
2.
Dev Growth Differ ; 64(7): 409-416, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36053973

RESUMO

The stimulated by retinoic acid gene 8 (STRA8)/MEIOSIN complex and polycomb repressive complex (PRC) 1.6, a PRC1 subtype, are believed to be positive and negative regulators of meiotic onset, respectively. During meiotic initiation, the transcription repressive activity of PRC1.6 must be attenuated so that meiosis-related genes can be effectively activated by the STRA8/MEIOSIN complex. However, the molecular mechanisms that control the impairment of PRC1.6 function remain unclear. We recently demonstrated that the Mga gene, which encodes a scaffolding component of PRC1.6, produces variant mRNA by alternative splicing specifically during meiosis. Furthermore, the anomalous MGA protein encoded by the variant mRNA bears an intrinsic ability to function as a dominant negative regulator against the construction of PRC1.6 and is therefore assumed to be, at least in part, involved in impairment of the complex. Therefore, to unequivocally evaluate the physiological significance of Mga variant mRNA production in gametogenesis, we examined the consequences of a genetic manipulation that renders mice unable to produce Mga variant mRNA. Our data revealed that mutant mice were equivalent to wild-type mice in terms of viability and fertility. Our detailed examination of spermatogenesis also revealed that this genetic alteration is not associated with any apparent abnormalities in testis size, spermatogenic cycle, timing of meiotic onset, or marker gene expression of spermatogonia and spermatocytes. Taken together, these data indicate that the production of germ cell-specific Mga variant mRNA is dispensable not only for viability but also for gametogenesis.


Assuntos
Processamento Alternativo , Células Germinativas , Processamento Alternativo/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fertilidade , Células Germinativas/metabolismo , Masculino , Meiose/genética , Camundongos , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espermatogênese/genética , Tretinoína/metabolismo
3.
Stem Cells ; 39(11): 1435-1446, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34224650

RESUMO

Although the physiological meaning of the high potential of mouse embryonic stem cells (ESCs) for meiotic entry is not understood, a rigid safeguarding system is required to prevent ectopic onset of meiosis. PRC1.6, a non-canonical PRC1, is known for its suppression of precocious and ectopic meiotic onset in germ cells and ESCs, respectively. MGA, a scaffolding component of PRC1.6, bears two distinct DNA-binding domains termed bHLHZ and T-box. However, it is unclear how this feature contributes to the functions of PRC1.6. Here, we demonstrated that both domains repress distinct sets of genes in murine ESCs, but substantial numbers of meiosis-related genes are included in both gene sets. In addition, our data demonstrated that bHLHZ is crucially involved in repressing the expression of Meiosin, which plays essential roles in meiotic entry with Stra8, revealing at least part of the molecular mechanisms that link negative and positive regulation of meiotic onset.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Meiose , Células-Tronco Embrionárias Murinas , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Células Germinativas , Meiose/genética , Camundongos
4.
Genome Res ; 28(6): 846-858, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29728365

RESUMO

In mouse embryonic stem cells (mESCs), the expression of provirus and endogenous retroelements is epigenetically repressed. Although many cellular factors involved in retroelement silencing have been identified, the complete molecular mechanism remains elusive. In this study, we performed a genome-wide CRISPR screen to advance our understanding of retroelement silencing in mESCs. The Moloney murine leukemia virus (MLV)-based retroviral vector MSCV-GFP, which is repressed by the SETDB1/TRIM28 pathway in mESCs, was used as a reporter provirus, and we identified more than 80 genes involved in this process. In particular, ATF7IP and the BAF complex components are linked with the repression of most of the SETDB1 targets. We characterized two factors, MORC2A and RESF1, of which RESF1 is a novel molecule in retroelement silencing. Although both factors are recruited to repress provirus, their roles in repression are different. MORC2A appears to function dependent on repressive epigenetic modifications, while RESF1 regulates repressive epigenetic modifications associated with SETDB1. Our genome-wide CRISPR screen cataloged genes which function at different levels in silencing of SETDB1-target retroelements and provides a useful resource for further molecular studies.


Assuntos
Epigênese Genética , Histona-Lisina N-Metiltransferase/genética , Proteínas Repressoras/genética , Retroelementos/genética , Fatores de Transcrição/genética , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Inativação Gênica , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Células-Tronco Embrionárias Murinas/virologia
5.
Dev Growth Differ ; 61(4): 283-293, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30919971

RESUMO

It is thought that the spleen contains stem cells that differentiate into somatic cells other than immune cells. We investigated the presence of these hypothetical splenic cells with stem cell characteristics and identified adherent cells forming densely-packed colonies (Splenic Adherent Colony-forming Cell; SACC) in the spleen. Splenic Adherent Colony-forming Cell was positive for alkaline phosphatase staining and stage-specific embryonic antigen (SSEA)-1 antigen. However, the self-renewal properties of SACCs were limited because they stopped cell proliferation once colonies visible to the naked eye were formed. Gene expression analyses by semi-quantitative RT-PCR revealed the significant expression of c-Myc and Klf4, whereas faint or no expression was evident for Nanog, Oct3/4, and Sox2. Global expression analyses by DNA microarray and subsequent gene ontology analyses revealed that the expression levels of genes related to the immune system were significantly lower in SACCs than in control splenic cells. In contrast, genes unrelated to the immune system, such as those involved in cell adhesion and axon guidance, were relatively highly expressed in SACCs compared with control splenic cells. Taken together, we identified a novel cell type residing in the spleen that is different from the hypothetical splenic stem cell, but which bears some, but not all, characteristics that represent an undifferentiated state.


Assuntos
Adesão Celular , Baço/citologia , Fosfatase Alcalina/análise , Animais , Proliferação de Células , Fator 4 Semelhante a Kruppel , Antígenos CD15/análise , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Baço/imunologia , Baço/metabolismo
6.
Stem Cells ; 36(9): 1355-1367, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29761578

RESUMO

Embryonic stem cells (ESCs) exhibit two salient features beneficial for regenerative medicine: unlimited self-renewal and pluripotency. Methyl-CpG-binding domain protein 3 (Mbd3), a scaffolding component of the nucleosome remodeling deacetylase complex, is a specific regulator of pluripotency, as ESCs lacking Mbd3 are defective for lineage commitment potential but retain normal self-renewal properties. However, functional similarities and dissimilarities among the three Mbd3 isoforms (a, b, and c) have not been intensively explored. Herein, we demonstrated that Mbd3c, which lacks an entire portion of the MBD domain, exerted equivalent activity for counteracting the defective lineage commitment potential of Mbd3-knockout ESCs. Our analyses also revealed that the coiled-coil domain common to all three MBD3 isoforms, but not the MBD domain, plays a crucial role in this activity. Mechanistically, our data demonstrate that the activity of the coiled-coil domain is exerted, at least in part, through recruitment of polycomb repressive complex 2 to a subset of genes linked to development and organogenesis, thus establishing stable transcriptional repression. Stem Cells 2018;36:1355-1367.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/citologia , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Camundongos , Domínios Proteicos , Isoformas de Proteínas , Fatores de Transcrição/química , Fatores de Transcrição/genética
7.
PLoS Genet ; 12(1): e1005679, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26741492

RESUMO

Mitochondrial disorders have the highest incidence among congenital metabolic disorders characterized by biochemical respiratory chain complex deficiencies. It occurs at a rate of 1 in 5,000 births, and has phenotypic and genetic heterogeneity. Mutations in about 1,500 nuclear encoded mitochondrial proteins may cause mitochondrial dysfunction of energy production and mitochondrial disorders. More than 250 genes that cause mitochondrial disorders have been reported to date. However exact genetic diagnosis for patients still remained largely unknown. To reveal this heterogeneity, we performed comprehensive genomic analyses for 142 patients with childhood-onset mitochondrial respiratory chain complex deficiencies. The approach includes whole mtDNA and exome analyses using high-throughput sequencing, and chromosomal aberration analyses using high-density oligonucleotide arrays. We identified 37 novel mutations in known mitochondrial disease genes and 3 mitochondria-related genes (MRPS23, QRSL1, and PNPLA4) as novel causative genes. We also identified 2 genes known to cause monogenic diseases (MECP2 and TNNI3) and 3 chromosomal aberrations (6q24.3-q25.1, 17p12, and 22q11.21) as causes in this cohort. Our approaches enhance the ability to identify pathogenic gene mutations in patients with biochemically defined mitochondrial respiratory chain complex deficiencies in clinical settings. They also underscore clinical and genetic heterogeneity and will improve patient care of this complex disorder.


Assuntos
Exoma/genética , Heterogeneidade Genética , Mitocôndrias/genética , Doenças Mitocondriais/genética , Adolescente , Criança , Pré-Escolar , Aberrações Cromossômicas , DNA Mitocondrial/genética , Feminino , Fibroblastos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mutação INDEL/genética , Lactente , Recém-Nascido , Masculino , Mitocôndrias/patologia , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/patologia , Polimorfismo de Nucleotídeo Único/genética
8.
Dev Growth Differ ; 59(2): 61-69, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28220481

RESUMO

Meiosis is a central event of sexual reproduction. Like somatic cells, germ cells conduct mitosis to increase their cell number, but unlike somatic cells, germ cells switch their cell division mode from mitosis to meiosis at a certain point in gametogenesis. However, the molecular basis of this switch remains elusive. In this review article, we give an overview of the onset of mammalian meiosis, including our recent finding that MYC Associated Factor X (MAX) prevents ectopic and precocious meiosis in embryonic stem cells (ESCs) and germ cells, respectively. We present a hypothetical model of a MAX-centered molecular network that regulates meiotic entry in mammals and propose that inducible Max knockout ESCs provide an excellent platform for exploring the molecular mechanisms of meiosis initiation, while excluding other aspects of gametogenesis.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Células-Tronco Embrionárias/metabolismo , Células Germinativas/metabolismo , Meiose/genética , Animais , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Células-Tronco Embrionárias/citologia , Gametogênese/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/citologia , Camundongos Knockout , Modelos Genéticos , Pesquisa/tendências
9.
Dev Growth Differ ; 59(8): 639-647, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28967672

RESUMO

The Oct4 gene is a master regulator of the pluripotent properties of embryonic stem cells (ESCs). Recently, Oct4 loci were shown to frequently localize in close proximity to one another during the early stage of cellular differentiation, implicating this event as an important prerequisite step for ESCs to exert their full differentiation potential. Although the differentiation capacity of embryonal carcinoma cells (ECCs), such as F9 and P19 ECC lines, is severely restricted compared with ESCs, ECCs bear a highly similar expression profile to that of ESCs including expression of Oct4 and other pluripotency marker genes. Therefore, we examined whether allelic pairing of Oct4 loci also occurs during differentiation of F9 and P19 ECCs. Our data clearly demonstrate that this event is only observed within ESCs, but not ECCs, subjected to induction of differentiation, indicating transient allelic pairing of Oct4 loci as a specific feature of pluripotent ESCs. Moreover, our data revealed that this pairing did not occur broadly across chromosome 17, which carries the Oct4 gene, but occurred locally between Oct4 loci, suggesting that Oct4 loci somehow exert a driving force for their allelic pairing.


Assuntos
Diferenciação Celular , Cromossomos Humanos Par 17 , Loci Gênicos , Células-Tronco Embrionárias Humanas/metabolismo , Fator 3 de Transcrição de Octâmero , Alelos , Linhagem Celular , Cromossomos Humanos Par 17/genética , Cromossomos Humanos Par 17/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Humanos , Fator 3 de Transcrição de Octâmero/biossíntese , Fator 3 de Transcrição de Octâmero/genética
10.
Stem Cells ; 34(2): 322-33, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26523946

RESUMO

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.


Assuntos
Técnicas de Reprogramação Celular/métodos , Proteínas de Ligação a DNA , Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Complexo Repressor Polycomb 2 , Receptores de Estrogênio , Fatores de Transcrição , Animais , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Fator 4 Semelhante a Kruppel , Camundongos , Complexo Repressor Polycomb 2/biossíntese , Complexo Repressor Polycomb 2/genética , Proteínas de Ligação a RNA , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
11.
Stem Cells ; 33(4): 1089-101, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25522312

RESUMO

Nucleostemin (NS) is a nucleolar GTP-binding protein that is involved in a plethora of functions including ribosomal biogenesis and maintenance of telomere integrity. In addition to its expression in cancerous cells, the NS gene is expressed in stem cells including embryonic stem cells (ESCs). Previous knockdown and knockout studies have demonstrated that NS is important to preserve the self-renewality and high expression levels of pluripotency marker genes in ESCs. Here, we found that forced expression of Nanog or Esrrb, but not other pluripotency factors, resulted in the dispensability of NS expression in ESCs. However, the detrimental phenotypes of ESCs associated with ablation of NS expression were not mitigated by forced expression of Rad51 or a nucleolar localization-defective NS mutant that counteracts the damage associated with loss of NS expression in other NS-expressing cells such as neural stem/progenitor cells. Thus, our results indicate that NS participates in preservation of the viability and integrity of ESCs, which is distinct from that in other NS-expressing cells.


Assuntos
Proteínas de Transporte/biossíntese , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas Nucleares/biossíntese , Receptores de Estrogênio/biossíntese , Animais , Proteínas de Ligação ao GTP , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Proteína Homeobox Nanog , Proteínas de Ligação a RNA
12.
Stem Cells ; 33(3): 713-25, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25385436

RESUMO

c-Myc and phosphatidylinositol 3-OH kinase (PI3K) both participate in diverse cellular processes, including cell cycle control and tumorigenic transformation. They also contribute to preserving embryonic stem cell (ESC) characteristics. However, in spite of the vast knowledge, the molecular relationship between c-Myc and PI3K in ESCs is not known. Herein, we demonstrate that c-Myc and PI3K function cooperatively but independently to support ESC self-renewal when murine ESCs are cultured under conventional culture condition. Interestingly, culture of ESCs in 2i-condition including a GSK3ß and MEK inhibitor renders both PI3K and Myc signaling dispensable for the maintenance of pluripotent properties. These results suggest that the requirement for an oncogenic proliferation-dependent mechanism sustained by Myc and PI3K is context dependent and that the 2i-condition liberates ESCs from the dependence of this mechanism.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/biossíntese , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Sistema de Sinalização das MAP Quinases , Camundongos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator 2 Relacionado a NF-E2/biossíntese , Fator 2 Relacionado a NF-E2/genética , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética
13.
Proc Natl Acad Sci U S A ; 110(16): 6412-7, 2013 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-23550161

RESUMO

Transcription factors (TFs) are able to regulate differentiation-related processes, including dedifferentiation and direct conversion, through the regulation of cell type-specific transcriptional profiles. However, the functional interactions between the TFs regulating different transcriptional profiles are not well understood. Here, we show that the TFs capable of inducing cell type-specific transcriptional profiles prevent the dedifferentiation induced by TFs for pluripotency. Of the large number of TFs expressed in a neural-lineage cell line, we identified a subset of TFs that, when overexpressed, strongly interfered with the dedifferentiation triggered by the procedure to generate induced pluripotent stem cells. This interference occurred through a maintenance mechanism of the cell type-specific transcriptional profile. Strikingly, the maintenance activity of the interfering TF set was strong enough to induce the cell line-specific transcriptional profile when overexpressed in a heterologous cell type. In addition, the TFs that interfered with dedifferentiation in hepatic-lineage cells involved TFs with known induction activity for hepatic-lineage cells. Our results suggest that dedifferentiation suppresses a cell type-specific transcriptional profile, which is primarily maintained by a small subset of TFs capable of inducing direct conversion. We anticipate that this functional correlation might be applicable in various cell types and might facilitate the identification of TFs with induction activity in efforts to understand differentiation.


Assuntos
Desdiferenciação Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Neurônios/metabolismo , Células-Tronco Pluripotentes/fisiologia , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Primers do DNA/genética , Perfilação da Expressão Gênica , Hepatócitos/citologia , Camundongos , Microscopia Eletrônica de Transmissão , Neurônios/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Plasmídeos/genética , RNA Interferente Pequeno/genética
14.
Biochem Biophys Res Commun ; 455(3-4): 347-52, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25446088

RESUMO

Fibrodysplasia ossificans progressiva (FOP) is a genetic disorder characterized by heterotopic endochondral ossification in soft tissue. A mutation in the bone morphogenetic protein (BMP) receptor ALK2, R206H, has been identified in patients with typical FOP. In the present study, we established murine embryonic stem (ES) cells that express wild-type human ALK2 or typical mutant human ALK2 [ALK2(R206H)] under the control of the Tet-Off system. Although wild-type ALK2 and mutant ALK2(R206H) were expressed in response to a withdrawal of doxycycline (Dox), BMP signaling was activated only in the mutant ALK2(R206H)-expressing cells without the addition of exogenous BMPs. The Dox-dependent induction of BMP signaling was blocked by a specific kinase inhibitor of the BMP receptor. The mutant ALK2(R206H)-carrying cells showed Dox-regulated chondrogenesis in vitro, which occurred in co-operation with transforming growth factor-ß1 (TGF-ß1). Overall, our ES cells are useful for studying the molecular mechanisms of heterotopic ossification in FOP in vitro and for developing novel inhibitors of chondrogenesis induced by mutant ALK2(R206H) associated with FOP.


Assuntos
Receptores de Ativinas Tipo I/genética , Condrogênese , Células-Tronco Embrionárias/citologia , Proteínas Mutantes/genética , Miosite Ossificante/genética , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular , Condrócitos/citologia , Modelos Animais de Doenças , Doxiciclina/química , Humanos , Imuno-Histoquímica , Camundongos , Mutação , Miosite Ossificante/metabolismo , Transdução de Sinais
15.
Am J Pathol ; 183(2): 592-603, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23885716

RESUMO

Nucleostemin (NS) is a nucleolar GTP-binding protein that is involved in ribosomal biogenesis and protection of telomeres. We investigated the expression of NS in human germ cell tumors and its function in a mouse germ cell tumor model. NS was abundantly expressed in undifferentiated, but not differentiated, types of human testicular germ cell tumors. NS was expressed concomitantly with OCT3/4, a critical regulator of the undifferentiated status of pluripotent stem cells in primordial germ cells and embryonal carcinomas. To investigate the roles of NS in tumor growth in vivo, we used a mouse teratoma model. Analysis of teratomas derived from embryonic stem cells in which the NS promoter drives GFP expression showed that cells highly expressing NS were actively proliferating and exhibited the characteristics of tumor-initiating cells, including the ability to initiate and propagate tumor cells in vivo. NS-expressing cells exhibited higher levels of GTP than non-NS-expressing cells. Because NS protein is stabilized by intracellular GTP, metabolic changes may contribute to abundant NS expression in the undifferentiated cells. OCT3/4 deficiency in teratomas led to loss of NS expression, resulting in growth retardation. Finally, we found that teratomas deficient in NS lost their undifferentiated characteristics, resulting in defective tumor proliferation. These data indicate that abundant expression of NS supports the undifferentiated properties of germ cell tumors.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Nucleares/metabolismo , Teratoma/metabolismo , Neoplasias Testiculares/metabolismo , Animais , Proliferação de Células , Modelos Animais de Doenças , Proteínas de Ligação ao GTP , Células Germinativas/patologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas de Ligação a RNA , Teratoma/patologia , Neoplasias Testiculares/patologia , Células Tumorais Cultivadas
16.
Nat Cell Biol ; 9(6): 625-35, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17515932

RESUMO

The pluripotency of embryonic stem (ES) cells is thought to be maintained by a few key transcription factors, including Oct3/4 and Sox2. The function of Oct3/4 in ES cells has been extensively characterized, but that of Sox2 has yet to be determined. Sox2 can act synergistically with Oct3/4 in vitro to activate Oct-Sox enhancers, which regulate the expression of pluripotent stem cell-specific genes, including Nanog, Oct3/4 and Sox2 itself. These findings suggest that Sox2 is required by ES cells for its Oct-Sox enhancer activity. Using inducible Sox2-null mouse ES cells, we show that Sox2 is dispensable for the activation of these Oct-Sox enhancers. In contrast, we demonstrate that Sox2 is necessary for regulating multiple transcription factors that affect Oct3/4 expression and that the forced expression of Oct3/4 rescues the pluripotency of Sox2-null ES cells. These results indicate that the essential function of Sox2 is to stabilize ES cells in a pluripotent state by maintaining the requisite level of Oct3/4 expression.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Células-Tronco Pluripotentes/metabolismo , Transativadores/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Proteínas de Ligação a DNA/genética , Desenvolvimento Embrionário/fisiologia , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Fatores de Transcrição SOXB1 , Transativadores/genética , Fatores de Transcrição/genética , Ativação Transcricional/genética , Regulação para Cima/genética
17.
Sci Rep ; 14(1): 5236, 2024 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-38433229

RESUMO

Meiosis is a specialized type of cell division that occurs physiologically only in germ cells. We previously demonstrated that MYC-associated factor X (MAX) blocks the ectopic onset of meiosis in embryonic and germline stem cells in culture systems. Here, we investigated the Max gene's role in mouse primordial germ cells. Although Max is generally ubiquitously expressed, we revealed that sexually undifferentiated male and female germ cells had abundant MAX protein because of their higher Max gene expression than somatic cells. Moreover, our data revealed that this high MAX protein level in female germ cells declined significantly around physiological meiotic onset. Max disruption in sexually undifferentiated germ cells led to ectopic and precocious expression of meiosis-related genes, including Meiosin, the gatekeeper of meiotic onset, in both male and female germ cells. However, Max-null male and female germ cells did not complete the entire meiotic process, but stalled during its early stages and were eventually eliminated by apoptosis. Additionally, our meta-analyses identified a regulatory region that supports the high Max expression in sexually undifferentiated male and female germ cells. These results indicate the strong connection between the Max gene and physiological onset of meiosis in vivo through dynamic alteration of its expression.


Assuntos
Fator X , Meiose , Animais , Feminino , Masculino , Camundongos , Apoptose , Pontos de Checagem do Ciclo Celular , Células Germinativas , Meiose/genética
18.
PLoS One ; 19(3): e0298264, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38547201

RESUMO

Although sevoflurane is one of the most commonly used inhalational anesthetic agents, the popularity of desflurane is increasing to a level similar to that of sevoflurane. Inhalational anesthesia generally activates and represses the expression of genes related to xenobiotic metabolism and immune response, respectively. However, there has been no comprehensive comparison of the effects of sevoflurane and desflurane on the expression of these genes. Thus, we used a next-generation sequencing method to compare alterations in the global gene expression profiles in the livers of rats subjected to inhalational anesthesia by sevoflurane or desflurane. Our bioinformatics analyses revealed that sevoflurane and, to a greater extent, desflurane significantly activated genes related to xenobiotic metabolism. Our analyses also revealed that both anesthetic agents, especially sevoflurane, downregulated many genes related to immune response.


Assuntos
Anestésicos Inalatórios , Isoflurano , Éteres Metílicos , Animais , Ratos , Sevoflurano/farmacologia , Desflurano , Isoflurano/farmacologia , Éteres Metílicos/farmacologia , Transcriptoma , Xenobióticos , Anestésicos Inalatórios/farmacologia , Anestesia por Inalação
19.
Stem Cells ; 30(8): 1634-44, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22696478

RESUMO

c-Myc participates in diverse cellular processes including cell cycle control, tumorigenic transformation, and reprogramming of somatic cells to induced pluripotent cells. c-Myc is also an important regulator of self-renewal and pluripotency of embryonic stem cells (ESCs). We recently demonstrated that loss of the Max gene, encoding the best characterized partner for all Myc family proteins, causes loss of the pluripotent state and extensive cell death in ESCs strictly in this order. However, the mechanisms and molecules that are responsible for these phenotypes remain largely obscure. Here, we show that Sirt1, p53, and p38(MAPK) are crucially involved in the detrimental phenotype of Max-null ESCs. Moreover, our analyses revealed that these proteins are involved at varying levels to one another in the hierarchy of the pathway leading to cell death in Max-null ESCs.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/biossíntese , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Sirtuína 1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Antioxidantes/farmacologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Morte Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Doxiciclina/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Fenótipo , Células-Tronco Pluripotentes/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética , Transfecção , Proteína Supressora de Tumor p53/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética
20.
Clin Exp Dent Res ; 9(4): 711-720, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37272305

RESUMO

OBJECTIVES: In recent years, an increase in oral cancer among elderly nonsmokers has been noted. The aim of this study was to identify novel oncogenes in oral cancer in older nonsmokers. MATERIAL AND METHODS: Whole-exome sequencing (WES) data from 324 oral cancer patients were obtained from The Cancer Genome Atlas. Single nucleotide variants (SNVs) and insertions/deletions (INDELs) were extracted from the WES data of older patients. Fisher's exact test was performed to determine the specificity of variants in these genes. Finally, SNVs and INDELs were identified by target enrichment sequencing. RESULTS: Gene ontology analysis of 112 genes with significant SNVs or INDELs in nonsmokers revealed that nonsynonymous SNVs in HECTD4 were significantly more frequent in nonsmokers than in smokers by target enrichment sequencing (p = .02). CONCLUSIONS: Further investigation of the function of HECTD4 variants as oncogenes in older nonsmokers is warranted.


Assuntos
Exoma , Neoplasias Bucais , Humanos , Idoso , não Fumantes , Polimorfismo de Nucleotídeo Único , Oncogenes/genética , Neoplasias Bucais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA