Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Biochem Biophys Res Commun ; 591: 50-53, 2022 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-34999253

RESUMEN

Cryopreservation of embryos is a useful method for stably preserving various strains for a long time, and the cryopreserved embryos can be used at any time by simple warming. However, the viability of cryopreserved embryos, particularly vitrification at an early stage, is low compared to that of fresh embryos. As the warming process during vitrification is known to affect the survivability and subsequent development of embryos, the present study aimed to examine the viability and subsequent development of vitrified early-stage mouse embryos after warming at different temperatures. The survival rate of pronuclear and 2-cell stage embryos warmed at 60 °C (97% and 88%, respectively) was significantly higher than that of the embryos warmed at 37 °C (46% and 48%, respectively). The pronuclear and 2-cell stage embryos warmed at 60 °C (86% and 100%) showed better development to the blastocyst stage than the embryos warmed at 37 °C (72% and 84%, respectively). The development of offspring of the surviving embryos was similar at both the warming temperatures. These results showed that the survivability and subsequent development of vitrified early-stage mouse embryos were obviously increased upon rapid warming. This improved warming process could be helpful for the maintenance and reproduction of genetic resources.


Asunto(s)
Criopreservación , Embrión de Mamíferos/fisiología , Temperatura , Vitrificación , Animales , Crioprotectores/farmacología , Ratones Endogámicos ICR
2.
Int Immunol ; 31(5): 335-347, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30726915

RESUMEN

Ten-eleven translocation (TET) proteins regulate DNA methylation and gene expression by converting 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). Although Tet2/Tet3 deficiency has been reported to lead to myeloid cell, B-cell and invariant natural killer T (iNKT) cell malignancy, the effect of TET on regulatory T cells (Tregs) has not been elucidated. We found that Tet2/Tet3 deficiency in Tregs led to lethal hyperproliferation of CD4+Foxp3+ T cells in the spleen and mesenteric lymph nodes after 5 months of age. Additionally, in aged Treg-specific Tet2/Tet3-deficient mice, serum IgG1, IgG3, IgM and IgE levels were markedly elevated. High IL-17 expression was observed in both Foxp3+ and Fopx3- CD4+ T cells, and adoptive transfer of Tet2/Tet3-deficient Tregs into lymphopenic mice inhibited Foxp3 expression and caused conversion into IL-17-producing cells. However, the conserved non-coding DNA sequence-2 (CNS2) region of the Foxp3 gene locus, which has been shown to be particularly important for stable Foxp3 expression, was only partly methylated. We identified novel TET-dependent demethylation sites in the Foxp3 upstream enhancer, which may contribute to stable Foxp3 expression. Together, these data indicate that Tet2 and Tet3 are involved in Treg stability and immune homeostasis in mice.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Dioxigenasas/inmunología , Factores de Transcripción Forkhead/metabolismo , Interleucina-17/biosíntesis , Proteínas Proto-Oncogénicas/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Animales , Proliferación Celular , Interleucina-17/inmunología , Ratones , Ratones Endogámicos C57BL
3.
Int Immunol ; 29(8): 365-375, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29048538

RESUMEN

Since induced regulatory T cells (iTregs) can be produced in a large quantity in vitro, these cells are expected to be clinically useful to induce immunological tolerance in various immunological diseases. Foxp3 (Forkhead box P3) expression in iTregs is, however, unstable due to the lack of demethylation of the CpG island in the conserved non-coding sequence 2 (CNS2) of the Foxp3 locus. To facilitate the demethylation of CNS2, we over-expressed the catalytic domain (CD) of the ten-eleven translocation (TET) protein, which catalyzes the steps of the iterative demethylation of 5-methylcytosine. TET-CD over-expression in iTregs resulted in partial demethylation of CNS2 and stable Foxp3 expression. We also discovered that TET expression was enhanced under low oxygen (5%) culture conditions, which facilitated CNS2 DNA demethylation and stabilization of Foxp3 expression in a TET2- and TET3-dependent manner. In combination with vitamin C treatment, which has been reported to enhance TET catalytic activity, iTregs generated under low oxygen conditions retained more stable Foxp3 expression in vitro and in vivo and exhibited stronger suppression activity in a colitis model compared with untreated iTregs. Our data indicate that the induction and activation of TET enzymes in iTregs would be an effective method for Treg-mediated adoptive immunotherapy.


Asunto(s)
Colitis/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead/metabolismo , Inmunoterapia Adoptiva/métodos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Ácido Ascórbico/administración & dosificación , Colitis/inducido químicamente , Secuencia Conservada , Islas de CpG/genética , Desmetilación , Dioxigenasas , Inducción Enzimática , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Humanos , Hipoxia , Ratones , Subgrupos de Linfocitos T/trasplante , Linfocitos T Reguladores/trasplante
4.
Proc Natl Acad Sci U S A ; 112(47): 14641-6, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26546670

RESUMEN

Substantial proportions of mammalian genomes comprise repetitive elements including endogenous retrotransposons. Although these play diverse roles during development, their appropriate silencing is critically important in maintaining genomic integrity in the host cells. The major mechanism for retrotransposon silencing is DNA methylation, but the wave of global DNA demethylation that occurs after fertilization renders preimplantation embryos exceptionally hypomethylated. Here, we show that hypomethylated preimplantation mouse embryos are protected from retrotransposons by repressive histone modifications mediated by the histone chaperone chromatin assembly factor 1 (CAF-1). We found that knockdown of CAF-1 with specific siRNA injections resulted in significant up-regulation of the retrotransposons long interspersed nuclear element 1, short interspersed nuclear element B2, and intracisternal A particle at the morula stage. Concomitantly, increased histone H2AX phosphorylation and developmental arrest of the majority (>95%) of embryos were observed. The latter was caused at least in part by derepression of retrotransposons, as treatment with reverse transcriptase inhibitors rescued some embryos. Importantly, ChIP analysis revealed that CAF-1 mediated the replacement of H3.3 with H3.1/3.2 at the retrotransposon regions. This replacement was associated with deposition of repressive histone marks, including trimethylation of histone H3 on lysine 9 (H3K9me3), H3K9me2, H3K27me3, and H4K20me3. Among them, H4K20me3 and H3K9me3 seemed to play predominant roles in retrotransposon silencing, as assessed by knockdown of specific histone methyltransferases and forced expression of unmethylatable mutants of H3.1K9 and H4K20. Our data thus indicate that CAF-1 is an essential guardian of the genome in preimplantation mouse embryos by deposition of repressive histone modifications via histone variant replacement.


Asunto(s)
Blastocisto/metabolismo , Factor 1 de Ensamblaje de la Cromatina/metabolismo , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Retroelementos/genética , Animales , Blastocisto/efectos de los fármacos , Núcleo Celular/metabolismo , Inmunoprecipitación de Cromatina , Femenino , Técnicas de Silenciamiento del Gen , Genes Dominantes , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Lisina/metabolismo , Masculino , Metilación/efectos de los fármacos , Ratones , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Mutación/genética , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología
5.
Genes Dev ; 24(5): 432-7, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20194436

RESUMEN

Methylation of histone H3 Lys 9 and Lys 27 (H3K9 and H3K27) is associated with transcriptional silencing. Here we show that KDM7, a JmjC domain-containing protein, catalyzes demethylation of both mono- or dimethylated H3K9 and H3K27. Inhibition of KDM7 orthologs in zebrafish resulted in developmental brain defects. KDM7 interacts with the follistatin gene locus, and KDM7 depletion in mammalian neuronal cells suppressed follistatin gene transcription in association with increased levels of dimethylated H3K9 and H3K27. Our findings identify KDM7 as a dual demethylase for H3K9 and H3K27 that functions as an eraser of silencing marks on chromatin during brain development.


Asunto(s)
Encéfalo/embriología , Encéfalo/enzimología , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Células Cultivadas , Embrión no Mamífero/embriología , Embrión no Mamífero/enzimología , Folistatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Neuronas/metabolismo , Pez Cebra/embriología
6.
Nature ; 439(7078): 811-6, 2006 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-16362057

RESUMEN

Covalent modification of histones has an important role in regulating chromatin dynamics and transcription. Whereas most covalent histone modifications are reversible, until recently it was unknown whether methyl groups could be actively removed from histones. Using a biochemical assay coupled with chromatography, we have purified a novel JmjC domain-containing protein, JHDM1 (JmjC domain-containing histone demethylase 1), that specifically demethylates histone H3 at lysine 36 (H3-K36). In the presence of Fe(ii) and alpha-ketoglutarate, JHDM1 demethylates H3-methyl-K36 and generates formaldehyde and succinate. Overexpression of JHDM1 reduced the level of dimethyl-H3-K36 (H3K36me2) in vivo. The demethylase activity of the JmjC domain-containing proteins is conserved, as a JHDM1 homologue in Saccharomyces cerevisiae also has H3-K36 demethylase activity. Thus, we identify the JmjC domain as a novel demethylase signature motif and uncover a protein demethylation mechanism that is conserved from yeast to human.


Asunto(s)
Histonas/metabolismo , Oxidorreductasas N-Desmetilantes/química , Oxidorreductasas N-Desmetilantes/metabolismo , Secuencias de Aminoácidos , Extractos Celulares , Secuencia Conservada , Proteínas F-Box , Formaldehído/metabolismo , Células HeLa , Histonas/química , Humanos , Histona Demetilasas con Dominio de Jumonji , Metilación , Oxidorreductasas N-Desmetilantes/clasificación , Oxidorreductasas N-Desmetilantes/genética , Estructura Terciaria de Proteína , Saccharomyces cerevisiae , Especificidad por Sustrato , Ácido Succínico/metabolismo
7.
Arthritis Res Ther ; 24(1): 222, 2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-36114544

RESUMEN

BACKGROUND: Rheumatoid arthritis (RA) patients present with abnormal methylation patterns in their fibroblast-like synoviocytes (FLS). Given that DNA demethylation is critical for producing DNA methylation patterns, we hypothesized that DNA demethylation may facilitate RA progression. Therefore, we designed this study to examine the role of DNA dioxygenase family, Ten-Eleven translocation (TET1/2/3), in the pathological process of RA. METHODS: Synovial tissues and FLS were obtained from patients with RA and Osteoarthritis. K/BxN serum-induced arthritis was induced in Wild-type (WT) and TET3 heterozygous-deficient (TET3+/-) C57BL/6 mice. RESULTS: We found that both TET3 and 5-hydroxymethylcytosine (5hmC) were upregulated in synovitis tissues from RA patients and confirmed this upregulation in the cultured FLS derived from synovitis tissues. Tumor necrosis factor α (TNFα) upregulated TET3 and 5hmC levels in cultured FLS, and the stimulated FLS exhibited high cell mobility with increased transcription of cellular migration-related factors such as C-X-C motif chemokine ligand 8 (CXCL8) and C-C motif chemokine ligand 2 (CCL2) in a TET3-dependent manner. In addition, TET3 haploinsufficiency lowered RA progression in a mouse model of serum-induced arthritis. CONCLUSIONS: Based on these findings, we can assume that TET3-mediated DNA demethylation acts as an epigenetic regulator of RA progression.


Asunto(s)
Artritis Reumatoide , Dioxigenasas/metabolismo , Sinovitis , Animales , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Quimiocinas , ADN , Dioxigenasas/genética , Ligandos , Ratones , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa
8.
Sci Rep ; 11(1): 11167, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34045607

RESUMEN

In multicellular organisms, oocytes and sperm undergo fusion during fertilization and the resulting zygote gives rise to a new individual. The ability of zygotes to produce a fully formed individual from a single cell when placed in a supportive environment is known as totipotency. Given that totipotent cells are the source of all multicellular organisms, a better understanding of totipotency may have a wide-ranging impact on biology. The precise delineation of totipotent cells in mammals has remained elusive, however, although zygotes and single blastomeres of embryos at the two-cell stage have been thought to be the only totipotent cells in mice. We now show that a single blastomere of two- or four-cell mouse embryos can give rise to a fertile adult when placed in a uterus, even though blastomere isolation disturbs the transcriptome of derived embryos. Single blastomeres isolated from embryos at the eight-cell or morula stages and cultured in vitro manifested pronounced defects in the formation of epiblast and primitive endoderm by the inner cell mass and in the development of blastocysts, respectively. Our results thus indicate that totipotency of mouse zygotes extends to single blastomeres of embryos at the four-cell stage.


Asunto(s)
Blastómeros/citología , Embrión de Mamíferos/citología , Desarrollo Embrionario/fisiología , Células Madre Totipotentes/citología , Cigoto/citología , Animales , Blastocisto/citología , Técnicas de Cultivo de Embriones , Ratones
9.
J Cell Biochem ; 104(1): 176-88, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18004740

RESUMEN

Strong activation of the ERK signal is required for hepatocyte growth factor (HGF) to inhibit proliferation of the human hepatocellular carcinoma cell line HepG2. However, it is still to be elucidated whether the activation alone is sufficient to induce the inhibitory effect. In this study, we constructed HepG2 cell clones expressing a high level of epidermal growth factor receptor (EGFR), and examined the effect of the strong activation of ERK on the proliferation of the cell clones. EGF treatment of the cell clones induced strong activation of ERK similar to HGF treatment, but did not inhibit cell proliferation. HGF treatment of the cell clones up-regulated the expression of a Cdk inhibitor p16(INK4a), which has previously been shown to be required to inhibit the proliferation of HepG2 cells, but EGF treatment did not. Furthermore, EGF treatment of the cell clones did not induce the up-regulation of another Cdk inhibitor p21(CIP1), whereas HGF treatment did. Knockdown of p21 by siRNA restored the proliferation of HepG2 cells inhibited by HGF, and restored Cdk2 activity suppressed in HGF-treated HepG2 cells. These results suggest that strong activation of ERK alone is not sufficient, and some other pathway(s), which is activated through the HGF receptor but not through EGFR, is also required to induce the up-regulation of p16 and p21 expression, and also suggest that in addition to the up-regulated expression of p16, that of p21 contributes to the suppression of Cdk2 activity leading to the inhibition of proliferation of HGF-treated HepG2 cells.


Asunto(s)
Carcinoma Hepatocelular/patología , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor de Crecimiento de Hepatocito/fisiología , Línea Celular Tumoral , Receptores ErbB , Humanos , Transducción de Señal , Regulación hacia Arriba
10.
Sci Rep ; 5: 15876, 2015 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-26507142

RESUMEN

The development of multicellular organisms is accompanied by reprogramming of the epigenome in specific cells, with the epigenome of most cell types becoming fixed after differentiation. Genome-wide reprogramming of DNA methylation occurs in primordial germ cells and in fertilized eggs during mammalian embryogenesis. The 5-methylcytosine (5mC) content of DNA thus undergoes a marked decrease in the paternal pronucleus of mammalian zygotes. This loss of DNA methylation has been thought to be mediated by an active demethylation mechanism independent of replication and to be required for development. TET3-mediated sequential oxidation of 5mC has recently been shown to contribute to the genome-wide loss of 5mC in the paternal pronucleus of mouse zygotes. We now show that TET3 localizes not only to the paternal pronucleus but also to the maternal pronucleus and oxidizes both paternal and maternal DNA in mouse zygotes, although these phenomena are less pronounced in the female pronucleus. Genetic ablation of TET3 in oocytes had no significant effect on oocyte development, maturation, or fertilization or on pregnancy, but it resulted in neonatal sublethality. Our results thus indicate that zygotic 5mC oxidation mediated by maternal TET3 is required for neonatal growth but is not essential for development.


Asunto(s)
Proteínas de Unión al ADN/genética , Embrión de Mamíferos/embriología , Embrión de Mamíferos/fisiología , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Proteínas Proto-Oncogénicas/genética , Cigoto/enzimología , Cigoto/crecimiento & desarrollo , 5-Metilcitosina/metabolismo , Animales , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Metilación de ADN/genética , Replicación del ADN/genética , Dioxigenasas , Embrión de Mamíferos/metabolismo , Femenino , Fertilización/genética , Fertilización/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Oocitos/enzimología , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Embarazo , Cigoto/metabolismo
11.
J Biochem ; 136(5): 701-9, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15632311

RESUMEN

Hepatocyte growth factor (HGF) induces growth stimulation of a variety of cell types, but it also induces growth inhibition of several types of tumor cell lines. We previously investigated the intracellular signaling pathway involved in the antiproliferative effect of HGF on the human hepatocellular carcinoma cell line HepG2. The results suggested that the HGF-induced proliferation inhibition is caused by cell cycle arrest, which results from the retinoblastoma tumor suppressor gene product pRb being maintained in its active hypophosphorylated form via a high-intensity ERK signal. In this study, we examined the molecular mechanism of the HGF-induced cell cycle arrest in HepG2 cells. Cyclin A/Cdk2 complexes phosphorylated serine residues on pRb crucial for the G1 to S phase transition in proliferating HepG2 cells, and HGF treatment inhibited the phosphorylation. The expression of cyclin A was decreased and the expression of a Cdk inhibitor p21(Cip1) was increased in HGF-treated HepG2 cells, and these changes were prevented by pretreatment with a low concentration of a MEK inhibitor. These results suggest that the decrease in cyclin A expression and increase in p21(Cip1) expression through a high-intensity ERK signal by HGF lead to suppression of the phosphorylation of pRb by Cdk2, which contributes to the cell cycle arrest at G1 in HepG2 cells by HGF. Furthermore, the expression of E2F-1, a member of the E2F transcription factor family, was decreased in HGF-treated HepG2 cells, suggesting that the decrease in E2F-1 expression may also contribute to the cell cycle arrest at G1.


Asunto(s)
Quinasas CDC2-CDC28/metabolismo , Carcinoma Hepatocelular/metabolismo , Ciclo Celular/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Neoplasias Hepáticas/metabolismo , Mitógenos/farmacología , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina A/biosíntesis , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Flavonoides/farmacología , Fase G1/efectos de los fármacos , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Fase S/efectos de los fármacos , Factores de Transcripción/biosíntesis , Factores de Transcripción/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1
12.
J Biochem ; 151(3): 229-46, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22247561

RESUMEN

Methylation of DNA and histones in chromatin has been implicated in numerous biological processes. For many years, methylation has been recognized as static and stable modification, as compared with other covalent modifications of chromatin. Recently, however, several mechanisms have been demonstrated to be involved in demethylation of chromatin, suggesting that chromatin methylation is more dynamically regulated. One chemical reaction that mediates demethylation of both DNA and histones is hydroxylation, catalysed by Fe(II) and α-ketoglutarate (KG)-dependent hydroxylase/dioxygenase. Given that methylation of chromatin is an important epigenetic mark involved in fundamental biological processes such as cell fate determination, understanding how chromatin methylation is dynamically regulated has implications for human diseases and regenerative medicine.


Asunto(s)
Cromatina/metabolismo , Hidroxilación/fisiología , 5-Metilcitosina/metabolismo , Animales , Citosina/análogos & derivados , Citosina/metabolismo , Metilación de ADN/fisiología , Humanos , Metilación
13.
Cold Spring Harb Protoc ; 2010(10): pdb.prot5512, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20889709

RESUMEN

Post-translational modifications of histones play an important role in regulating chromatin dynamics and function. One of the modifications, methylation, occurs on both lysine and arginine residues, and methylation status defines the epigenetic program of a cell by determining chromatin structure and thereby regulating DNA-dependent processes such as transcription. Until recently, histone methylation was considered to be irreversible. However, the discovery of histone demethylases revealed that histone methylation is more dynamic than previously recognized. This protocol describes two different in vitro histone demethylase enzyme reactions and three different methods for measuring histone demethylase activity. The first reaction (type I) uses the Fe(II)- and α-ketoglutarate-dependent dioxygenase family of histone demethylase (represented by JmjC domain-containing histone demethylase [JHDM]); the second (type II) is for the flavin adenine dinucleotide (FAD)-dependent amine oxidase family (represented by lysine-specific demethylase 1 [LSD1]). Histone demethylase activity can then be detected by measuring the release of radiolabeled formaldehyde from (3)H-labeled methylated histone substrates, by monitoring the change in methylation levels of histone substrates by immunoblotting with site-specific methylhistone antibodies, or by using mass spectrometry to detect reductions in histone peptide masses that correspond to methyl groups. These assays can be applied to a wide range of histone demethylase studies, including the measurement of histone demethylase activity in tissue and cell lysates, identification of novel histone demethylases, and screening for inhibitors of histone demethylases.


Asunto(s)
Bioensayo/métodos , Histona Demetilasas/metabolismo , Histona Demetilasas/análisis
14.
Nat Cell Biol ; 11(2): 172-82, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19151705

RESUMEN

The chromodomain helicase DNA-binding (CHD) family of enzymes is thought to regulate gene expression, but their role in the regulation of specific genes has been unclear. Here we show that CHD8 is expressed at a high level during early embryogenesis and prevents apoptosis mediated by the tumour suppressor protein p53. CHD8 was found to bind to p53 and to suppress its transactivation activity. CHD8 promoted the association of p53 and histone H1, forming a trimeric complex on chromatin that was required for inhibition of p53-dependent transactivation and apoptosis. Depletion of CHD8 or histone H1 resulted in p53 activation and apoptosis. Furthermore, Chd8(-/-) mice died early during embryogenesis, manifesting widespread apoptosis, whereas deletion of p53 ameliorated this developmental arrest. These observations reveal a mode of p53 regulation mediated by CHD8, which may set a threshold for induction of apoptosis during early embryogenesis by counteracting p53 function through recruitment of histone H1.


Asunto(s)
Apoptosis/genética , Cadherinas/genética , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica/genética , Histonas/genética , Proteína p53 Supresora de Tumor/genética , Animales , Línea Celular , Regulación hacia Abajo/genética , Células HeLa , Humanos , Sustancias Macromoleculares/metabolismo , Ratones , Ratones Noqueados , Unión Proteica/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
15.
Nat Struct Mol Biol ; 15(11): 1169-75, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18836456

RESUMEN

The Ink4a-Arf-Ink4b locus has a crucial role in both cellular senescence and tumorigenesis. JmjC domain-containing histone demethylase 1b (Jhdm1b, also known as Kdm2b and Fbxl10), the mammalian paralog of the histone demethylase Jhdm1a (also known as Kdm2a and Fbxl11), has been implicated in cell-cycle regulation and tumorigenesis. In this report, we show that Jhdm1b is a histone H3 lysine 36 (H3K36) demethylase. Knockdown of Jhdm1b in primary mouse embryonic fibroblasts inhibits cell proliferation and induces cellular senescence in a pRb- and p53 pathway-dependent manner. Notably, the effect of Jhdm1b on cell proliferation and cellular senescence is mediated through derepression of p15(Ink4b), as loss of p15(Ink4b) function rescues cell-proliferation defects in Jhdm1b-knockdown cells. Chromatin immunoprecipitation on ectopically expressed Jhdm1b demonstrates that Jhdm1b targets the p15(Ink4b) locus and regulates its expression in an enzymatic activity-dependent manner. Alteration of Jhdm1b level affects Ras-induced neoplastic transformation. Collectively, our results indicate that Jhdm1b is an H3K36 demethylase that regulates cell proliferation and senescence through p15(Ink4b).


Asunto(s)
Proliferación Celular , Senescencia Celular/fisiología , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas F-Box/metabolismo , Animales , Transformación Celular Neoplásica , Células Cultivadas , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas F-Box/genética , Fibroblastos/citología , Fibroblastos/fisiología , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji , Ratones , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , Proteínas ras/genética , Proteínas ras/metabolismo
16.
Methods ; 40(4): 318-26, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17101443

RESUMEN

Posttranslational histone modifications play an important role in regulating chromatin dynamics and function. One of the modifications, methylation, occurs on both lysine and arginine residues and participates in diverse range of biological processes including heterochromatin formation, X-chromosome inactivation, and transcriptional regulation. While acetylation, phosphorylation, and ubiquitylation are dynamically regulated by enzymes that catalyze the addition and removal of a particular modification, enzymes that are capable of removing methyl groups were not known until recently. Thus far, two families of histone demethylases with distinct cofactor requirements and reaction mechanisms have been identified. One is the FAD (flavin adenine dinucleotide)-dependent amine oxidase family LSD1 (lysine-specific demethylase), the other is the Fe(II) and alpha-KG (alpha-ketoglutarate)-dependent dioxygenase family JHDM (JmjC domain-containing histone demethylase). Identification and characterization of these histone demethylases is an important step towards understanding both the function and regulation of histone methylation. Here, we describe assays currently used for measuring histone demethylase activity and chromatography strategies used in purifying histone demethylases from HeLa cells.


Asunto(s)
Histonas/metabolismo , Complejos Multiproteicos/aislamiento & purificación , Oxidorreductasas N-Desmetilantes/análisis , Oxidorreductasas N-Desmetilantes/aislamiento & purificación , Cromatografía/métodos , Proteínas F-Box , Células HeLa , Histona Demetilasas , Humanos , Histona Demetilasas con Dominio de Jumonji , Metilación
17.
Cancer Sci ; 97(12): 1343-50, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17032311

RESUMEN

The association of hepatocyte growth factor (HGF) with its high-affinity receptor (c-Met) has been shown to induce mitogenesis, motogenesis and morphogenesis in a variety of cell types. Various point mutations in c-Met have been identified in hereditary and sporadic papillary renal carcinomas as well as in other carcinomas. In the present study, we examined the effects of c-Met point mutations on the morphology of a porcine aortic endothelial (PAE) cell line. When cultured in three-dimensional collagen gel, PAE cells formed branching tubule structures, and HGF treatment caused breakdown of the structures and induced a scattered morphology. The exogenous expression of c-Met point mutants inhibited the formation of tubules. HGF treatment induced the formation of tubules by PAE cells expressing some c-Met mutants, but it induced the scattering of PAE cells expressing other c-Met mutants. The presence of a low concentration of a mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK) inhibitor cancelled the inhibitory effect of the c-Met point mutations on the formation of tubules. These results suggest that c-Met point mutations affect the extracellular signal-regulated kinase (ERK) signaling required for the formation of tubules by PAE cells, and HGF binding changes the conformation of c-Met mutants, leading to the different signals required for formation of tubules and cell scattering.


Asunto(s)
Aorta/crecimiento & desarrollo , Endotelio Vascular/citología , Proteínas Proto-Oncogénicas c-met/farmacología , Animales , Aorta/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Quinasas Quinasa Quinasa PAM/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Transducción de Señal , Porcinos
18.
Cell ; 125(3): 483-95, 2006 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-16603237

RESUMEN

Covalent modification of histones plays an important role in regulating chromatin dynamics and transcription. Histone methylation was thought to be an irreversible modification until recently. Using a biochemical assay coupled with chromatography, we have purified a JmjC domain-containing protein, JHDM2A, which specifically demethylates mono- and dimethyl-H3K9. Similar to JHDM1, JHDM2A-mediated histone demethylation requires cofactors Fe(II) and alpha-ketoglutarate. Mutational studies indicate that a JmjC domain and a zinc finger present in JHDM2A are required for its enzymatic activity. Overexpression of JHDM2A greatly reduced the H3K9 methylation level in vivo. Knockdown of JHDM2A results in an increase in the dimethyl-K9 levels at the promoter region of a subset of genes concomitant with decrease in their expression. Finally, JHDM2A exhibits hormone-dependent recruitment to androgen-receptor target genes, resulting in H3K9 demethylation and transcriptional activation. Thus, our work identifies a histone demethylase and links its function to hormone-dependent transcriptional activation.


Asunto(s)
Histonas/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo , Receptores Androgénicos/metabolismo , Transactivadores/metabolismo , Activación Transcripcional/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Dominio Catalítico/genética , Chlorocebus aethiops , Regulación hacia Abajo/fisiología , Hormonas Esteroides Gonadales/metabolismo , Células HeLa , Histonas/genética , Humanos , Hierro/metabolismo , Histona Demetilasas con Dominio de Jumonji , Ácidos Cetoglutáricos/metabolismo , Ratones , Datos de Secuencia Molecular , Oxidorreductasas N-Desmetilantes/genética , Oxidorreductasas N-Desmetilantes/aislamiento & purificación , Regiones Promotoras Genéticas/fisiología , Estructura Terciaria de Proteína/genética , Transactivadores/genética , Transactivadores/aislamiento & purificación
19.
Mol Cell ; 20(6): 845-54, 2005 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-16359901

RESUMEN

Polycomb group (PcG) proteins exist in at least two biochemically distinct protein complexes, the EED-EZH2 complex and the PRC1 complex, that respectively possess H3-K27 methyltransferase and H2A-K119 ubiquitin E3 ligase activities. How the enzymatic activities are regulated and what their role is in Hox gene silencing are not clear. Here, we demonstrate that Bmi-1 and Ring1A, two components of the PRC1 complex, play important roles in H2A ubiquitylation and Hox gene silencing. We show that both proteins positively regulate H2A ubiquitylation. Chromatin immunoprecipitation (ChIP) assays demonstrate that Bmi-1 and other components of the two PcG complexes bind to the promoter of HoxC13. Knockout Bmi-1 results in significant loss of H2A ubiquitylation and upregulation of Hoxc13 expression, whereas EZH2-mediated H3-K27 methylation is not affected. Our results suggest that EZH2-mediated H3-K27 methylation functions upstream of PRC1 and establishes a critical role for Bmi-1 and Ring1A in H2A ubiquitylation and Hox gene silencing.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Silenciador del Gen , Genes Homeobox , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina/metabolismo , Animales , Línea Celular , Cromatina/metabolismo , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Histonas/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Noqueados , Complejos Multiproteicos , Proteínas Nucleares/genética , Complejo Represivo Polycomb 1 , Complejo Represivo Polycomb 2 , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Ubiquitina-Proteína Ligasas/metabolismo
20.
J Biol Chem ; 280(36): 31548-56, 2005 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-16014626

RESUMEN

Hepatocyte growth factor (HGF) has an anti-proliferative effect on many types of tumor cell lines and tumors in vivo. We found previously that inhibition of HGF-induced proliferation in HepG2 hepatoma cells is caused by cell cycle arrest at G1 through a high intensity ERK signal, which represses Cdk2 activity. To examine further the mechanisms of G1 arrest by HGF, we analyzed the Cdk inhibitor p16(INK4a), which has an anti-proliferative function through cell cycle arrest at G1. We found that HGF treatment drastically increased endogenous p16 levels. Knockdown of p16 with small interfering RNA reversed the arrest, indicating that the induction of p16 is required for G1 arrest by HGF. Analysis of the promoter of the human p16 gene identified the proximal Ets-binding site as a responsive element for HGF, and this responded to the high intensity ERK signal. HGF treatment of the cells led to a redistribution of p21(CIP1) and p27(KIP1) from Cdk4 to Cdk2. The redistribution was blocked by the knockdown of p16 with small interfering RNA, which restored the Cdk2 activity repressed by HGF, demonstrating the requirement of p16 induction for the redistribution and eventual repression of Cdk2 activity. Our results reveal a signaling pathway for G1 arrest induced by HGF.


Asunto(s)
Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Fase G1/fisiología , Factor de Crecimiento de Hepatocito/fisiología , Neoplasias Hepáticas/patología , Proteínas Supresoras de Tumor/metabolismo , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/metabolismo , Ciclo Celular/fisiología , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/metabolismo , Regulación hacia Arriba/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA