Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
2.
Nat Immunol ; 21(8): 950-961, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32572241

RESUMO

A contribution of epigenetic modifications to B cell tolerance has been proposed but not directly tested. Here we report that deficiency of ten-eleven translocation (Tet) DNA demethylase family members Tet2 and Tet3 in B cells led to hyperactivation of B and T cells, autoantibody production and lupus-like disease in mice. Mechanistically, in the absence of Tet2 and Tet3, downregulation of CD86, which normally occurs following chronic exposure of self-reactive B cells to self-antigen, did not take place. The importance of dysregulated CD86 expression in Tet2- and Tet3-deficient B cells was further demonstrated by the restriction, albeit not complete, on aberrant T and B cell activation following anti-CD86 blockade. Tet2- and Tet3-deficient B cells had decreased accumulation of histone deacetylase 1 (HDAC1) and HDAC2 at the Cd86 locus. Thus, our findings suggest that Tet2- and Tet3-mediated chromatin modification participates in repression of CD86 on chronically stimulated self-reactive B cells, which contributes, at least in part, to preventing autoimmunity.


Assuntos
Autoimunidade/imunologia , Linfócitos B/imunologia , Antígeno B7-2/imunologia , Proteínas de Ligação a DNA/imunologia , Dioxigenases/imunologia , Proteínas Proto-Oncogênicas/imunologia , Animais , Doenças Autoimunes/imunologia , Epigênese Genética/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
3.
Nat Immunol ; 15(6): 571-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24777532

RESUMO

Intestinal regulatory T cells (Treg cells) are necessary for the suppression of excessive immune responses to commensal bacteria. However, the molecular machinery that controls the homeostasis of intestinal Treg cells has remained largely unknown. Here we report that colonization of germ-free mice with gut microbiota upregulated expression of the DNA-methylation adaptor Uhrf1 in Treg cells. Mice with T cell-specific deficiency in Uhrf1 (Uhrf1(fl/fl)Cd4-Cre mice) showed defective proliferation and functional maturation of colonic Treg cells. Uhrf1 deficiency resulted in derepression of the gene (Cdkn1a) that encodes the cyclin-dependent kinase inhibitor p21 due to hypomethylation of its promoter region, which resulted in cell-cycle arrest of Treg cells. As a consequence, Uhrf1(fl/fl)Cd4-Cre mice spontaneously developed severe colitis. Thus, Uhrf1-dependent epigenetic silencing of Cdkn1a was required for the maintenance of gut immunological homeostasis. This mechanism enforces symbiotic host-microbe interactions without an inflammatory response.


Assuntos
Colite/imunologia , Colo/imunologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Epigênese Genética , Proteínas Nucleares/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Proteínas Estimuladoras de Ligação a CCAAT , Pontos de Checagem do Ciclo Celular , Proliferação de Células , Células Cultivadas , Clostridium/imunologia , Colite/genética , Colo/microbiologia , Metilação de DNA , Perfilação da Expressão Gênica , Interleucina-2 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microbiota/imunologia , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno , Simbiose/imunologia , Ubiquitina-Proteína Ligases , Regulação para Cima
4.
Development ; 148(10)2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33998651

RESUMO

Heterochromatin-related epigenetic mechanisms, such as DNA methylation, facilitate pairing of homologous chromosomes during the meiotic prophase of mammalian spermatogenesis. In pro-spermatogonia, de novo DNA methylation plays a key role in completing meiotic prophase and initiating meiotic division. However, the role of maintenance DNA methylation in the regulation of meiosis, especially in the adult, is not well understood. Here, we reveal that NP95 (also known as UHRF1) and DNMT1 - two essential proteins for maintenance DNA methylation - are co-expressed in spermatogonia and are necessary for meiosis in male germ cells. We find that Np95- or Dnmt1-deficient spermatocytes exhibit spermatogenic defects characterized by synaptic failure during meiotic prophase. In addition, assembly of pericentric heterochromatin clusters in early meiotic prophase, a phenomenon that is required for subsequent pairing of homologous chromosomes, is disrupted in both mutants. Based on these observations, we propose that DNA methylation, established in pre-meiotic spermatogonia, regulates synapsis of homologous chromosomes and, in turn, quality control of male germ cells. Maintenance DNA methylation, therefore, plays a role in ensuring faithful transmission of both genetic and epigenetic information to offspring.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Pareamento Cromossômico/genética , DNA (Citosina-5-)-Metiltransferase 1/genética , Metilação de DNA/genética , Espermatócitos/crescimento & desenvolvimento , Espermatogênese/genética , Ubiquitina-Proteína Ligases/genética , Células-Tronco Germinativas Adultas/citologia , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Epigênese Genética/genética , Heterocromatina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Espermatócitos/fisiologia , Espermatogênese/fisiologia , Ubiquitina-Proteína Ligases/metabolismo
5.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33893234

RESUMO

The stratum corneum (SC), the outermost epidermal layer, consists of nonviable anuclear keratinocytes, called corneocytes, which function as a protective barrier. The exact modes of cell death executed by keratinocytes of the upper stratum granulosum (SG1 cells) remain largely unknown. Here, using intravital imaging combined with intracellular Ca2+- and pH-responsive fluorescent probes, we aimed to dissect the SG1 death process in vivo. We found that SG1 cell death was preceded by prolonged (∼60 min) Ca2+ elevation and rapid induction of intracellular acidification. Once such intracellular ionic changes were initiated, they became sustained, irreversibly committing the SG1 cells to corneocyte conversion. Time-lapse imaging of isolated murine SG1 cells revealed that intracellular acidification was essential for the degradation of keratohyalin granules and nuclear DNA, phenomena specific to SC corneocyte formation. Furthermore, intravital imaging showed that the number of SG1 cells exhibiting Ca2+ elevation and the timing of intracellular acidification were both tightly regulated by the transient receptor potential cation channel V3. The functional activity of this protein was confirmed in isolated SG1 cells using whole-cell patch-clamp analysis. These findings provide a theoretical framework for improved understanding of the unique molecular mechanisms underlying keratinocyte-specific death mode, namely corneoptosis.


Assuntos
Morte Celular/fisiologia , Células Epidérmicas/metabolismo , Queratinócitos/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Diferenciação Celular , Epiderme/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Queratinócitos/fisiologia , Camundongos , Camundongos Transgênicos , Técnicas de Patch-Clamp/métodos , Pele
6.
Nature ; 548(7666): 219-223, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28746311

RESUMO

Concomitant activation of the Wnt pathway and suppression of Mapk signalling by two small molecule inhibitors (2i) in the presence of leukaemia inhibitory factor (LIF) (hereafter termed 2i/L) induces a naive state in mouse embryonic stem (ES) cells that resembles the inner cell mass (ICM) of the pre-implantation embryo. Since the ICM exists only transiently in vivo, it remains unclear how sustained propagation of naive ES cells in vitro affects their stability and functionality. Here we show that prolonged culture of male mouse ES cells in 2i/L results in irreversible epigenetic and genomic changes that impair their developmental potential. Furthermore, we find that female ES cells cultured in conventional serum plus LIF medium phenocopy male ES cells cultured in 2i/L. Mechanistically, we demonstrate that the inhibition of Mek1/2 is predominantly responsible for these effects, in part through the downregulation of DNA methyltransferases and their cofactors. Finally, we show that replacement of the Mek1/2 inhibitor with a Src inhibitor preserves the epigenetic and genomic integrity as well as the developmental potential of ES cells. Taken together, our data suggest that, although short-term suppression of Mek1/2 in ES cells helps to maintain an ICM-like epigenetic state, prolonged suppression results in irreversible changes that compromise their developmental potential.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Animais , Blastocisto , Instabilidade Cromossômica , Metilação de DNA , Feminino , Impressão Genômica , Cariotipagem , Masculino , Camundongos
7.
Glia ; 70(7): 1267-1288, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35262217

RESUMO

The human brain is a complex, three-dimensional structure. To better recapitulate brain complexity, recent efforts have focused on the development of human-specific midbrain organoids. Human iPSC-derived midbrain organoids consist of differentiated and functional neurons, which contain active synapses, as well as astrocytes and oligodendrocytes. However, the absence of microglia, with their ability to remodel neuronal networks and phagocytose apoptotic cells and debris, represents a major disadvantage for the current midbrain organoid systems. Additionally, neuroinflammation-related disease modeling is not possible in the absence of microglia. So far, no studies about the effects of human iPSC-derived microglia on midbrain organoid neural cells have been published. Here we describe an approach to derive microglia from human iPSCs and integrate them into iPSC-derived midbrain organoids. Using single nuclear RNA Sequencing, we provide a detailed characterization of microglia in midbrain organoids as well as the influence of their presence on the other cells of the organoids. Furthermore, we describe the effects that microglia have on cell death and oxidative stress-related gene expression. Finally, we show that microglia in midbrain organoids affect synaptic remodeling and increase neuronal excitability. Altogether, we show a more suitable system to further investigate brain development, as well as neurodegenerative diseases and neuroinflammation.


Assuntos
Células-Tronco Pluripotentes Induzidas , Organoides , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Mesencéfalo , Microglia/metabolismo , Neurogênese/genética , Organoides/metabolismo
8.
Development ; 145(19)2018 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-30190278

RESUMO

Suppression of Meis genes in the distal limb bud is required for proximal-distal (PD) specification of the forelimb. Polycomb group (PcG) factors play a role in downregulation of retinoic acid (RA)-related signals in the distal forelimb bud, causing Meis repression. It is, however, not known whether downregulation of RA-related signals and PcG-mediated proximal gene repression are functionally linked. Here, we reveal that PcG factors and RA-related signals antagonize each other to polarize Meis2 expression along the PD axis in mouse. Supported by mathematical modeling and simulation, we propose that PcG factors are required to adjust the threshold for RA-related signaling to regulate Meis2 expression. Finally, we show that a variant Polycomb repressive complex 1 (PRC1), incorporating PCGF3 and PCGF5, represses Meis2 expression in the distal limb bud. Taken together, we reveal a previously unknown link between PcG proteins and downregulation of RA-related signals to mediate the phase transition of Meis2 transcriptional status during forelimb patterning.


Assuntos
Membro Anterior/embriologia , Proteínas de Homeodomínio/metabolismo , Botões de Extremidades/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Tretinoína/metabolismo , Animais , Membro Anterior/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Loci Gênicos , Camundongos , Transdução de Sinais
9.
PLoS Genet ; 13(10): e1007042, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28976982

RESUMO

The methylation of cytosine at CG sites in the mammalian genome is dynamically reprogrammed during gametogenesis and preimplantation development. It was previously shown that oocyte-derived DNMT1 (a maintenance methyltransferase) is essential for maintaining and propagating CG methylation at imprinting control regions in preimplantation embryos. In mammalian somatic cells, hemimethylated-CG-binding protein UHRF1 plays a critical role in maintaining CG methylation by recruiting DNMT1 to hemimethylated CG sites. However, the role of UHRF1 in oogenesis and preimplantation development is unknown. In the present study, we show that UHRF1 is mainly, but not exclusively, localized in the cytoplasm of oocytes and preimplantation embryos. However, smaller amounts of UHRF1 existed in the nucleus, consistent with the expected role in DNA methylation. We then generated oocyte-specific Uhrf1 knockout (KO) mice and found that, although oogenesis was itself unaffected, a large proportion of the embryos derived from the KO oocytes died before reaching the blastocyst stage (a maternal effect). Whole genome bisulfite sequencing revealed that blastocysts derived from KO oocytes have a greatly reduced level of CG methylation, suggesting that maternal UHRF1 is essential for maintaining CG methylation, particularly at the imprinting control regions, in preimplantation embryos. Surprisingly, UHRF1 was also found to contribute to de novo CG and non-CG methylation during oocyte growth: in Uhrf1 KO oocytes, transcriptionally-inactive regions gained less methylation, while actively transcribed regions, including the imprinting control regions, were unaffected or only slightly affected. We also found that de novo methylation was defective during the late stage of oocyte growth. To the best of our knowledge, this is the first study to demonstrate the role of UHRF1 in de novo DNA methylation in vivo. Our study reveals multiple functions of UHRF1 during the global epigenetic reprogramming of oocytes and early embryos.


Assuntos
Blastocisto/metabolismo , Metilação de DNA , Proteínas Nucleares/metabolismo , Oócitos/metabolismo , Animais , Proteínas Estimuladoras de Ligação a CCAAT , Reprogramação Celular , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Desenvolvimento Embrionário , Epigênese Genética , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Oócitos/crescimento & desenvolvimento , Oogênese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Frações Subcelulares/metabolismo , Ubiquitina-Proteína Ligases
10.
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
11.
Nature ; 502(7470): 249-53, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-24013172

RESUMO

Faithful propagation of DNA methylation patterns during DNA replication is critical for maintaining cellular phenotypes of individual differentiated cells. Although it is well established that Uhrf1 (ubiquitin-like with PHD and ring finger domains 1; also known as Np95 and ICBP90) specifically binds to hemi-methylated DNA through its SRA (SET and RING finger associated) domain and has an essential role in maintenance of DNA methylation by recruiting Dnmt1 to hemi-methylated DNA sites, the mechanism by which Uhrf1 coordinates the maintenance of DNA methylation and DNA replication is largely unknown. Here we show that Uhrf1-dependent histone H3 ubiquitylation has a prerequisite role in the maintenance DNA methylation. Using Xenopus egg extracts, we successfully reproduce maintenance DNA methylation in vitro. Dnmt1 depletion results in a marked accumulation of Uhrf1-dependent ubiquitylation of histone H3 at lysine 23. Dnmt1 preferentially associates with ubiquitylated H3 in vitro though a region previously identified as a replication foci targeting sequence. The RING finger mutant of Uhrf1 fails to recruit Dnmt1 to DNA replication sites and maintain DNA methylation in mammalian cultured cells. Our findings represent the first evidence, to our knowledge, of the mechanistic link between DNA methylation and DNA replication through histone H3 ubiquitylation.


Assuntos
Metilação de DNA/fisiologia , Replicação do DNA/fisiologia , Histonas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/genética , Xenopus laevis/metabolismo , Animais , Linhagem Celular , Metilação de DNA/genética , Replicação do DNA/genética , Células HEK293 , Células HeLa , Humanos , Camundongos , Óvulo/química , Ligação Proteica , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Proteínas de Xenopus/genética
12.
Development ; 140(17): 3565-76, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23903187

RESUMO

Epigenetic modifications influence gene expression and chromatin remodeling. In embryonic pluripotent stem cells, these epigenetic modifications have been extensively characterized; by contrast, the epigenetic events of tissue-specific stem cells are poorly understood. Here, we define a new epigenetic shift that is crucial for differentiation of murine spermatogonia toward meiosis. We have exploited a property of incomplete cytokinesis, which causes male germ cells to form aligned chains of characteristic lengths, as they divide and differentiate. These chains revealed the stage of spermatogenesis, so the epigenetic differences of various stages could be characterized. Single, paired and medium chain-length spermatogonia not expressing Kit (a marker of differentiating spermatogonia) showed no expression of Dnmt3a2 and Dnmt3b (two de novo DNA methyltransferases); they also lacked the transcriptionally repressive histone modification H3K9me2. By contrast, spermatogonia consisting of ~8-16 chained cells with Kit expression dramatically upregulated Dnmt3a2/3b expression and also displayed increased H3K9me2 modification. To explore the function of these epigenetic changes in spermatogonia in vivo, the DNA methylation machinery was destabilized by ectopic Dnmt3b expression or Np95 ablation. Forced Dnmt3b expression induced expression of Kit; whereas ablation of Np95, which is essential for maintaining DNA methylation, interfered with differentiation and viability only after spermatogonia become Kit positive. These data suggest that the epigenetic status of spermatogonia shifts dramatically during the Kit-negative to Kit-positive transition. This shift might serve as a switch that determines whether spermatogonia self-renew or differentiate.


Assuntos
Diferenciação Celular/fisiologia , Epigênese Genética/fisiologia , Células Germinativas/fisiologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Espermatogênese/fisiologia , Espermatogônias/crescimento & desenvolvimento , Animais , Western Blotting , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/fisiologia , Primers do DNA/genética , Citometria de Fluxo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Espermatogônias/citologia , DNA Metiltransferase 3B
13.
PLoS Genet ; 9(11): e1003897, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24244179

RESUMO

Molecular mechanisms for the establishment of transcriptional memory are poorly understood. 5,6-dichloro-1-D-ribofuranosyl-benzimidazole (DRB) is a P-TEFb kinase inhibitor that artificially induces the poised RNA polymerase II (RNAPII), thereby manifesting intermediate steps for the establishment of transcriptional activation. Here, using genetics and DRB, we show that mammalian Absent, small, or homeotic discs 1-like (Ash1l), a member of the trithorax group proteins, methylates Lys36 of histone H3 to promote the establishment of Hox gene expression by counteracting Polycomb silencing. Importantly, we found that Ash1l-dependent Lys36 di-, tri-methylation of histone H3 in a coding region and exclusion of Polycomb group proteins occur independently of transcriptional elongation in embryonic stem (ES) cells, although both were previously thought to be consequences of transcription. Genome-wide analyses of histone H3 Lys36 methylation under DRB treatment have suggested that binding of the retinoic acid receptor (RAR) to a certain genomic region promotes trimethylation in the RAR-associated gene independent of its ongoing transcription. Moreover, DRB treatment unveils a parallel response between Lys36 methylation of histone H3 and occupancy of either Tip60 or Mof in a region-dependent manner. We also found that Brg1 is another key player involved in the response. Our results uncover a novel regulatory cascade orchestrated by Ash1l with RAR and provide insights into mechanisms underlying the establishment of the transcriptional activation that counteracts Polycomb silencing.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Proteínas do Grupo Polycomb/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Fatores de Elongação da Transcrição/genética , Animais , Cromatina/genética , Proteínas de Ligação a DNA/genética , Diclororribofuranosilbenzimidazol/farmacologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Inativação Gênica , Histona Acetiltransferases/metabolismo , Histona-Lisina N-Metiltransferase , Histonas/genética , Humanos , Lisina/genética , Metilação , Proteínas Nucleares/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Fator B de Elongação Transcricional Positiva/antagonistas & inibidores , Fator B de Elongação Transcricional Positiva/metabolismo , RNA Polimerase II/genética , Fatores de Transcrição/genética , Fatores de Elongação da Transcrição/metabolismo
14.
J Biol Chem ; 289(17): 11656-11666, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24584930

RESUMO

Histone variants play specific roles in maintenance and regulation of chromatin structures. H2ABbd, an H2A variant, possesses a highly divergent structure compared with canonical H2A and is highly expressed in postmeiotic germ cells, but its functions in the regulation of gene expression are largely unknown. In the present study, we investigated the cellular phenotype associated with enforced H2ABbd expression. Among H2A variants, H2ABbd specifically caused growth defect in human cells and induced apoptosis. H2ABbd expression resulted in degradation of inhibitor of κB-α and translocation of NF-κB into nuclei, indicating the activation of NF-κB. Intriguingly, NF-κB activity was essential for H2ABbd-induced apoptosis. H2ABbd overexpression resulted in DNA damage after release from G1/S, progressed through the S phase slowly, and induced apoptosis. Furthermore, gene expression microarray analysis revealed that expression of H2ABbd activates groups of genes involved in apoptosis and postmeiotic germ cell development, suggesting that H2ABbd might influence transcription. Taken together, our data suggest that H2ABbd may contribute to specific chromatin structures and promote NF-κB activation, which could in turn induce apoptosis in mammalian cells.


Assuntos
Apoptose/fisiologia , Histonas/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Replicação do DNA , Histonas/genética , Humanos , Reação em Cadeia da Polimerase em Tempo Real
15.
Biochem Biophys Res Commun ; 458(3): 470-475, 2015 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-25660450

RESUMO

Epigenetic modifications such as DNA methylation and histone H3 lysine 27 methylation (H3K27me) are repressive marks that silence gene expression. The M phase phosphoprotein (MPP8) associates with proteins involved in both DNA methylation and histone modifications, and therefore, is a potential candidate to mediate crosstalk between repressive epigenetic pathways. Here, by performing immunohistochemical analyses we demonstrate that MPP8 is expressed in the rodent testis, especially in spermatocytes, suggesting a role in spermatogenesis. Interestingly, we found that MPP8 physically interacts with PRC1 (Polycomb Repressive Complex 1) components which are known to possess essential function in testis development by modulating monoubiquitination of Histone H2A (uH2A) and trimethylation of Histone H3 Lysine 27 (H3K27me3) residues. Knockdown analysis of MPP8 in HeLa cells resulted in derepression of a set of genes that are normally expressed in spermatogonia, spermatids and mature sperm, thereby indicating a role for this molecule in silencing testis-related genes in somatic cells. In addition, depletion of MPP8 in murine ES cells specifically induced expression of genes involved in mesoderm differentiation, such as Cdx2 and Brachyury even in the presence of LIF, which implicated that MPP8 might be required to repress differentiation associated genes during early development. Taken together, our results indicate that MPP8 could have a role for silencing genes that are associated with differentiation of the testis and the mesoderm by interacting with epigenetic repressors modules such as the PRC1 complex.


Assuntos
Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Espermatogênese , Animais , Linhagem Celular , Metilação de DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Técnicas de Silenciamento de Genes , Células HeLa , Histonas/metabolismo , Humanos , Masculino , Camundongos , Fosfoproteínas/análise , Complexo Repressor Polycomb 1/análise , Mapas de Interação de Proteínas , Ratos Endogâmicos F344 , Espermatócitos/citologia , Espermatócitos/metabolismo , Ativação Transcricional
16.
PLoS Genet ; 8(7): e1002774, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22844243

RESUMO

Two distinct Polycomb complexes, PRC1 and PRC2, collaborate to maintain epigenetic repression of key developmental loci in embryonic stem cells (ESCs). PRC1 and PRC2 have histone modifying activities, catalyzing mono-ubiquitination of histone H2A (H2AK119u1) and trimethylation of H3 lysine 27 (H3K27me3), respectively. Compared to H3K27me3, localization and the role of H2AK119u1 are not fully understood in ESCs. Here we present genome-wide H2AK119u1 maps in ESCs and identify a group of genes at which H2AK119u1 is deposited in a Ring1-dependent manner. These genes are a distinctive subset of genes with H3K27me3 enrichment and are the central targets of Polycomb silencing that are required to maintain ESC identity. We further show that the H2A ubiquitination activity of PRC1 is dispensable for its target binding and its activity to compact chromatin at Hox loci, but is indispensable for efficient repression of target genes and thereby ESC maintenance. These data demonstrate that multiple effector mechanisms including H2A ubiquitination and chromatin compaction combine to mediate PRC1-dependent repression of genes that are crucial for the maintenance of ESC identity. Utilization of these diverse effector mechanisms might provide a means to maintain a repressive state that is robust yet highly responsive to developmental cues during ES cell self-renewal and differentiation.


Assuntos
Histonas , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ubiquitinação , Animais , Linhagem Celular , Cromatina/genética , Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/genética , Histonas/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/genética
17.
Development ; 138(19): 4207-17, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21896631

RESUMO

During meiosis, specific histone modifications at pericentric heterochromatin (PCH), especially histone H3 tri- and dimethylation at lysine 9 (H3K9me3 and H3K9me2, respectively), are required for proper chromosome interactions. However, the molecular mechanism by which H3K9 methylation mediates the synapsis is not yet understood. We have generated a Cbx3-deficient mouse line and performed comparative analysis on Suv39h1/h2-, G9a- and Cbx3-deficient spermatocytes. This study revealed that H3K9me2 at PCH depended on Suv39h1/h2-mediated H3K9me3 and its recognition by the Cbx3 gene product HP1γ. We further found that centromere clustering and synapsis were commonly affected in G9a- and Cbx3-deficient spermatocytes. These genetic observations suggest that HP1γ/G9a-dependent PCH-mediated centromere clustering is an axis for proper chromosome interactions during meiotic prophase. We propose that the role of the HP1γ/G9a axis is to retain centromeric regions of unpaired homologous chromosomes in close alignment and facilitate progression of their pairing in early meiotic prophase. This study also reveals considerable plasticity in the interplay between different histone modifications and suggests that such stepwise and dynamic epigenetic modifications may play a pivotal role in meiosis.


Assuntos
Proteínas Cromossômicas não Histona/genética , Pareamento Cromossômico , Histona-Lisina N-Metiltransferase/genética , Histonas/química , Meiose , Animais , Centrômero/ultraestrutura , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/fisiologia , Epigênese Genética , Feminino , Histona-Lisina N-Metiltransferase/fisiologia , Masculino , Metilação , Camundongos , Camundongos Knockout , Microscopia de Fluorescência/métodos , Modelos Genéticos , Espermatócitos/citologia
18.
Sci Rep ; 14(1): 14732, 2024 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-38926604

RESUMO

Excess amounts of histones in the cell induce mitotic chromosome loss and genomic instability, and are therefore detrimental to cell survival. In yeast, excess histones are degraded by the proteasome mediated via the DNA damage response factor Rad53. Histone expression, therefore, is tightly regulated at the protein level. Our understanding of the transcriptional regulation of histone genes is far from complete. In this study, we found that calcineurin inhibitor treatment increased histone protein levels, and that the transcription factor NFATc1 (nuclear factor of activated T cells 1) repressed histone transcription and acts downstream of the calcineurin. We further revealed that NFATc1 binds to the promoter regions of many histone genes and that histone transcription is downregulated in a manner dependent on intracellular calcium levels. Indeed, overexpression of histone H3 markedly inhibited cell proliferation. Taken together, these findings suggest that NFATc1 prevents the detrimental effects of histone H3 accumulation by inhibiting expression of histone at the transcriptional level.


Assuntos
Calcineurina , Histonas , Fatores de Transcrição NFATC , Fatores de Transcrição NFATC/metabolismo , Fatores de Transcrição NFATC/genética , Histonas/metabolismo , Calcineurina/metabolismo , Humanos , Proliferação de Células , Regulação da Expressão Gênica , Regiões Promotoras Genéticas , Transdução de Sinais , Transcrição Gênica , Cálcio/metabolismo
19.
Front Cell Dev Biol ; 12: 1410177, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38911025

RESUMO

Mammalian germ cells are derived from primordial germ cells (PGCs) and ensure species continuity through generations. Unlike irreversible committed mature germ cells, migratory PGCs exhibit a latent pluripotency characterized by the ability to derive embryonic germ cells (EGCs) and form teratoma. Here, we show that inhibition of p38 mitogen-activated protein kinase (MAPK) by chemical compounds in mouse migratory PGCs enables derivation of chemically induced Embryonic Germ-like Cells (cEGLCs) that do not require conventional growth factors like LIF and FGF2/Activin-A, and possess unique naïve pluripotent-like characteristics with epiblast features and chimera formation potential. Furthermore, cEGLCs are regulated by a unique PI3K-Akt signaling pathway, distinct from conventional naïve pluripotent stem cells described previously. Consistent with this notion, we show by performing ex vivo analysis that inhibition of p38 MAPK in organ culture supports the survival and proliferation of PGCs and also potentially reprograms PGCs to acquire indefinite proliferative capabilities, marking these cells as putative teratoma-producing cells. These findings highlight the utility of our ex vivo model in mimicking in vivo teratoma formation, thereby providing valuable insights into the cellular mechanisms underlying tumorigenesis. Taken together, our research underscores a key role of p38 MAPK in germ cell development, maintaining proper cell fate by preventing unscheduled pluripotency and teratoma formation with a balance between proliferation and differentiation.

20.
Nature ; 450(7171): 908-12, 2007 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-17994007

RESUMO

DNA methyltransferase (cytosine-5) 1 (Dnmt1) is the principal enzyme responsible for maintenance of CpG methylation and is essential for the regulation of gene expression, silencing of parasitic DNA elements, genomic imprinting and embryogenesis. Dnmt1 is needed in S phase to methylate newly replicated CpGs occurring opposite methylated ones on the mother strand of the DNA, which is essential for the epigenetic inheritance of methylation patterns in the genome. Despite an intrinsic affinity of Dnmt1 for such hemi-methylated DNA, the molecular mechanisms that ensure the correct loading of Dnmt1 onto newly replicated DNA in vivo are not understood. The Np95 (also known as Uhrf1 and ICBP90) protein binds methylated CpG through its SET and RING finger-associated (SRA) domain. Here we show that localization of mouse Np95 to replicating heterochromatin is dependent on the presence of hemi-methylated DNA. Np95 forms complexes with Dnmt1 and mediates the loading of Dnmt1 to replicating heterochromatic regions. By using Np95-deficient embryonic stem cells and embryos, we show that Np95 is essential in vivo to maintain global and local DNA methylation and to repress transcription of retrotransposons and imprinted genes. The link between hemi-methylated DNA, Np95 and Dnmt1 thus establishes key steps of the mechanism for epigenetic inheritance of DNA methylation.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA/metabolismo , Epigênese Genética , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Animais , Proteínas Estimuladoras de Ligação a CCAAT , Ilhas de CpG/genética , DNA/química , DNA (Citosina-5-)-Metiltransferase 1 , Replicação do DNA , Células-Tronco Embrionárias/metabolismo , Impressão Genômica , Células HeLa , Heterocromatina/genética , Heterocromatina/metabolismo , Humanos , Camundongos , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Estrutura Terciária de Proteína , Retroelementos/genética , Transcrição Gênica , Ubiquitina-Proteína Ligases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA