Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 36(27): 3934-3942, 2017 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-28192404

RESUMO

Despite extensive effort, there has been limited progress in the development of direct RAS inhibitors. Targeting isoprenylcysteine carboxylmethyltransferase (ICMT), a unique enzyme of RAS post-translational modification, represents a promising strategy to inhibit RAS function. However, there lacks direct genetic evidence on the role of ICMT in RAS-driven human cancer initiation and maintenance. Using CRISPR/Cas9 genome editing, we have created Icmt loss-of-function isogenic cell lines for both RAS-transformed human mammary epithelial cells (HME1) and human cancer cell lines MiaPaca-2 and MDA-MB-231 containing naturally occurring mutant KRAS. In both in vitro and in vivo tumorigenesis studies, Icmt loss-of-function abolishes the tumor initiation ability of all major isoforms of mutant RAS in HME1 cells, and the tumor maintenance capacity of MiaPaca-2 and MDA-MB-231 cells, establishing the critical role of ICMT in RAS-driven cancers.


Assuntos
Proteínas Metiltransferases/fisiologia , Proteínas ras/genética , Animais , Antineoplásicos Hormonais/farmacologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Humanos , Camundongos SCID , Mutação de Sentido Incorreto , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Tamoxifeno/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas ras/metabolismo
2.
Nucleic Acids Res ; 43(16): 7931-44, 2015 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-26206670

RESUMO

Recent studies have shown that homologous recombination (HR) requires chromatin repression as well as relaxation at DNA double strand breaks (DSBs). HP1 and SUV39H1/2 are repressive factors essential for HR. Here, we identify SETDB1 as an additional compacting factor promoting HR. Depletion of HP1, SUV39, SETDB1 or BRCA1 confer identical phenotypes. The repressive factors, like BRCA1, are dispensable for the initiation of resection but promote the extension step causing diminished RPA or RAD51 foci and HR in irradiated G2 cells. Depletion of the compacting factors does not inhibit BRCA1 recruitment but at 8 h post IR, BRCA1 foci are smaller and aberrantly positioned compared to control cells. BRCA1 promotes 53BP1 repositioning to the periphery of enlarged foci and formation of a devoid core with BRCA1 becoming enlarged and localized internally to 53BP1. Depletion of the compacting factors precludes these changes at irradiation-induced foci. Thus, the repressive factors are required for BRCA1 function in promoting the repositioning of 53BP1 during HR. Additionally, depletion of these repressive factors in undamaged cells causes diminished sister chromatid association at centromeric sequences. We propose a model for how these findings may be functionally linked.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Histona-Lisina N-Metiltransferase/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Metiltransferases/fisiologia , Proteínas Metiltransferases/fisiologia , Reparo de DNA por Recombinação , Proteínas Repressoras/fisiologia , Proteína BRCA1/metabolismo , Células Cultivadas , Cromátides , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Dano ao DNA , Reparo do DNA por Junção de Extremidades , Fase G2 , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Humanos , Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
3.
Nucleic Acids Res ; 42(22): 13545-56, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25183519

RESUMO

In mammals, RNA interference is primarily a post-transcriptional mechanism. Evidence has accumulated for additional role in transcriptional gene silencing (TGS) but the question for a good paradigm for small interfering antigene RNA (agRNA)-induced chromatin modification remains unanswered. Here, we show that SETDB1, a histone H3-lysine 9 (H3K9)-specific methyltransferase, cooperates with Argonaute-2 (AGO2) and plays an essential role in agRNA-induced TGS. The androgen receptor (AR) gene was transcriptionally silenced by agRNA targeted to its promoter, and we show that this repression was mitigated by knockdown of SETDB1 or AGO2. Chromatin immunoprecipitation demonstrated that agRNA-driven AGO2 was first targeted to the AR promoter, followed by SETDB1. SIN3A and HDAC1/2, the components of the SIN3-HDAC complex, immunoprecipitated with SETDB1, and localized at the agRNA-targeted promoter. Agreeing with the presence of SETDB1, trimethyl-H3K9 was enriched in the AR promoter. Both EZH2 and trimethyl-H3K27 were also present in the targeted locus; accordingly, EZH2 immunoprecipitated with SETDB1. DNA methylation level was not significantly changed, suggesting the absence of de novo methylating activity in agRNA-induced AR promoter. Our results demonstrate that SETDB1, together with AGO2, plays an essential role in TGS through recruiting chromatin remodeler and/or other modifiers, consequently creating a repressive chromatin milieu at the targeted promoter.


Assuntos
Proteínas Argonautas/metabolismo , Inativação Gênica , Proteínas Metiltransferases/metabolismo , Receptores Androgênicos/genética , Transcrição Gênica , Animais , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Camundongos , Células NIH 3T3 , Complexo Repressor Polycomb 2/metabolismo , Regiões Promotoras Genéticas , Proteínas Metiltransferases/fisiologia , RNA não Traduzido/análise
4.
Asian J Androl ; 16(2): 319-24, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24556744

RESUMO

SETDB1 has been established as an oncogene in a number of human carcinomas. The present study was to evaluate the expression of SETDB1 in prostate cancer (PCa) tissues and cells and to preliminarily investigate the role of SETDB1 in prostate tumorigenesis in vitro. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) were used to detect the expression of SETDB1 in PCa tissues, adjacent normal tissues, benign prostatic hyperplasia (BPH) tissues, PCa cell lines and normal prostate epithelial cells. The results suggested that SETDB1 was upregulated in human PCa tissues compared with normal tissues at the mRNA and protein levels. The role of SETDB1 in proliferation was analyzed with cell counting kit-8, colony-forming efficiency and flow cytometry assays. The results indicated that downregulation of SETDB1 by siRNA inhibited PCa cell growth, and induced G0/G1 cell cycle arrest. The PCa cell migration and invasion decreased by silcencing SETDB1 which were assessed by using in vitro scratch and transwell invasion assay respectively. Our data suggested that SETDB1 is overexpressed in human PCa. Silencing SETDB1 inhibited PCa cell proliferation, migration and invasion.


Assuntos
Invasividade Neoplásica , Metástase Neoplásica , Neoplasias da Próstata/patologia , Proteínas Metiltransferases/fisiologia , Sequência de Bases , Ciclo Celular , Primers do DNA , Inativação Gênica , Histona-Lisina N-Metiltransferase , Humanos , Masculino , Proteínas Metiltransferases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Neuromolecular Med ; 16(1): 70-82, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23943221

RESUMO

Posttranslational modifications of histones are considered as critical regulators of gene expression, playing significant role in the pathogenesis and progression of tumors. Trimethylation of histone 3 lysine 9 (H3K9me3), a repressed transcription mark, is mainly regulated by the histone lysine N-methyltransferases (HKMTs), SUV39H1 and SETDB1. The present study investigated the implication of these HKMTs in glioma progression. SUV39H1 and SETDB1 expression was upregulated in glioma cell lines (GOS-3, 1321N1, T98G, U87MG) and in glioma tissues compared to normal brain being positively correlated with grade and histological malignancy. Suppression by siRNA of the two HKMTs for 24 and 48 h resulted in significantly reduced proliferation of GOS-3 and T98G glioma cells with siSUV39H1 effects been most prominent. Furthermore, HKMTs knockdown-induced apoptosis with a high rate of apoptotic cells have been observed after siSUV39H1 and siSETDB1 for both cell lines. Additionally, suppression of the two HKMTs reduced cell migration and clonogenic ability of both glioma cell lines. Our results indicate overexpression of SETDB1 and SUV39H1 in gliomas. Treatments that alter HKMT expression affect the proliferative and apoptotic rates in glioma cells as well as their migratory and colony formation capacity. These data suggest that both HKMTs and especially SUV39H1 may serve as novel biomarkers for future therapeutic targeting of these tumors.


Assuntos
Astrocitoma/enzimologia , Neoplasias Encefálicas/enzimologia , Glioblastoma/enzimologia , Metiltransferases/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas Metiltransferases/fisiologia , Proteínas Repressoras/fisiologia , Apoptose/efeitos dos fármacos , Astrocitoma/patologia , Biomarcadores Tumorais , Neoplasias Encefálicas/patologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Indução Enzimática , Glioblastoma/patologia , Histona-Lisina N-Metiltransferase , Humanos , Lisina/química , Metilação , Metiltransferases/análise , Metiltransferases/antagonistas & inibidores , Gradação de Tumores , Proteínas de Neoplasias/análise , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas Metiltransferases/análise , Proteínas Metiltransferases/antagonistas & inibidores , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Proteínas Repressoras/análise , Proteínas Repressoras/antagonistas & inibidores , Ensaio Tumoral de Célula-Tronco , Regulação para Cima
6.
Clin Cancer Res ; 19(23): 6344-50, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23958745

RESUMO

The protein methyltransferases (PMT) constitute a large and important class of enzymes that catalyze site-specific methylation of lysine or arginine residues on histones and other proteins. Site-specific histone methylation is a critical component of chromatin regulation of gene transcription-a pathway that is often genetically altered in human cancers. Oncogenic alterations (e.g., mutations, chromosomal translocations, and others) of PMTs, or of associated proteins, have been found to confer unique dependencies of cancer cells on the activity of specific PMTs. Examples of potent, selective small-molecule inhibitors of specific PMTs are reviewed that have been shown to kill cancers cells bearing such oncogenic alterations, while having minimal effect on proliferation of nonaltered cells. Selective inhibitors of the PMTs, DOT1L and EZH2, have entered phase I clinical studies and additional examples of selective PMT inhibitors are likely to enter the clinic soon. The current state of efforts toward clinical testing of selective PMT inhibitors as personalized cancer therapeutics is reviewed here.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Proteínas Metiltransferases/fisiologia , Processamento de Proteína Pós-Traducional , Animais , Antineoplásicos/uso terapêutico , Domínio Catalítico , Ensaios Clínicos como Assunto , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Terapia de Alvo Molecular , Mutação , Neoplasias/enzimologia , Neoplasias/genética , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/química
7.
Mol Endocrinol ; 26(4): 583-97, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22361822

RESUMO

Regulation of adipose tissue formation by adipogenic-regulatory proteins has long been a topic of interest given the ever-increasing health concerns of obesity and type 2 diabetes in the general population. Differentiation of precursor cells into adipocytes involves a complex network of cofactors that facilitate the functions of transcriptional regulators from the CCATT/enhancer binding protein, and the peroxisome proliferator-activated receptor (PPAR) families. Many of these cofactors are enzymes that modulate the structure of chromatin by altering histone-DNA contacts in an ATP-dependent manner or by posttranslationally modifying the histone proteins. Here we report that inhibition of protein arginine methyltransferase 5 (Prmt5) expression in multiple cell culture models for adipogenesis prevented the activation of adipogenic genes. In contrast, overexpression of Prmt5 enhanced adipogenic gene expression and differentiation. Chromatin immunoprecipitation experiments indicated that Prmt5 binds to and dimethylates histones at adipogenic promoters. Furthermore, the presence of Prmt5 promoted the binding of ATP-dependent chromatin-remodeling enzymes and was required for the binding of PPARγ2 at PPARγ2-regulated promoters. The data indicate that Prmt5 acts as a coactivator for the activation of adipogenic gene expression and promotes adipogenic differentiation.


Assuntos
Adipogenia/genética , Expressão Gênica , PPAR gama/genética , Proteínas Metiltransferases/fisiologia , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Montagem e Desmontagem da Cromatina , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Regulação da Expressão Gênica , Histonas/metabolismo , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/metabolismo , Metilação , Camundongos , PPAR gama/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases , Técnicas de Cultura de Tecidos
8.
Mol Cell ; 43(4): 673-80, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21855805

RESUMO

Methylation of specific lysine residues in the C terminus of p53 is thought to govern p53-dependent transcription following genotoxic and oncogenic stress. In particular, Set7/9 (KMT7)-mediated monomethylation of human p53 at lysine 372 (p53K372me1) was suggested to be essential for p53 activation in human cell lines. This finding was confirmed in a Set7/9 knockout mouse model (Kurash et al., 2008). In an independent knockout mouse strain deficient in Set7/9, we have investigated its involvement in p53 regulation and find that cells from these mice are normal in their ability to induce p53-dependent transcription following genotoxic and oncogenic insults. Most importantly, we detect no impairment in canonical p53 functions in these mice, indicating that Set7/9-mediated methylation of p53 does not seem to represent a major regulatory event and does not appreciably control p53 activity in vivo.


Assuntos
Proteínas Metiltransferases/genética , Transcrição Gênica , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose/genética , Ciclo Celular , Senescência Celular/genética , Regulação da Expressão Gênica , Histona-Lisina N-Metiltransferase , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Metiltransferases/metabolismo , Proteínas Metiltransferases/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
9.
Mol Cell ; 43(4): 681-8, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21855806

RESUMO

p53 is the central regulator of cell fate following genotoxic stress and oncogene activation. Its activity is controlled by several posttranslational modifications. Originally defined as a critical layer of p53 regulation in human cell lines, p53 lysine methylation by Set7/9 (also called Setd7) was proposed to fulfill a similar function in vivo in the mouse, promoting p53 acetylation, stabilization, and activation upon DNA damage (Kurash et al., 2008). We tested the physiological relevance of this circuit in an independent Set7/9 knockout mouse strain. Deletion of Set7/9 had no effect on p53-dependent cell-cycle arrest or apoptosis following sublethal or lethal DNA damage induced by radiation or genotoxic agents. Set7/9 was also dispensable for p53 acetylation following irradiation. c-myc oncogene-induced apoptosis was also independent of Set7/9, and analysis of p53 target genes showed that Set7/9 is not required for the p53-dependent gene expression program. Our data indicate that Set7/9 is dispensable for p53 function in the mouse.


Assuntos
Dano ao DNA , Proteínas Metiltransferases/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Acetilação , Animais , Apoptose/genética , Ciclo Celular/genética , Deleção de Genes , Regulação da Expressão Gênica , Histona-Lisina N-Metiltransferase , Camundongos , Camundongos Knockout , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Proteína Supressora de Tumor p53/metabolismo
10.
Islets ; 3(2): 48-57, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21346419

RESUMO

Isoprenylcysteine carboxyl methyltransferase (ICMT) catalyzes the post-translational methylation of C-terminal cysteines of isoprenylated proteins, including small G-proteins and the γ-subunits of heterotrimeric G-proteins. It is widely felt that carboxymethylation promotes efficient membrane association of the methylated proteins and specific protein-protein interactions. In the current study, we tested the hypothesis that ICMT-mediated carboxymethylation of specific proteins (e.g., Rac1) plays a regulatory role in glucose-stimulated insulin secretion (GSIS). Western blot analysis indicated that lCMT is expressed and predominantly membrane associated in INS 832/13 ß-cells. siRNA-mediated knockdown of endogenous expression of ICMT markedly attenuated glucose, but not KCl-induced insulin secretion. These findings were further supported by pharmacological observations, which suggested a marked reduction in glucose-, but not KCl-stimulated insulin secretion by acetyl farnesyl cysteine (AFC), a selective inhibitor of ICMT. In addition, glucose-induced Rac1 activation, a hallmark signaling step involved in glucose-stimulated insulin secretion, was markedly inhibited following pharmacological (AFC) or molecular biological (siRNA-ICMT) inhibition of ICMT. Lastly, we also noticed a marked reduction in glucose-induced acute increase in the generation of reactive oxygen species in INS 832/13 cells pre-treated with AFC or transfected with siRNA-ICMT. Together, these data suggest that ICMT regulates glucose-induced Rac1 activation, generation of reactive oxygen species and insulin secretion in pancreatic ß-cells.


Assuntos
Glucose/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas Metiltransferases/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Retículo Endoplasmático/metabolismo , Secreção de Insulina , Cloreto de Potássio/farmacologia , RNA Interferente Pequeno/genética , Ratos
11.
Genes Dev ; 24(24): 2772-7, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21159818

RESUMO

Prmt5, an arginine methyltransferase, has multiple roles in germ cells, and possibly in pluripotency. Here we show that loss of Prmt5 function is early embryonic-lethal due to the abrogation of pluripotent cells in blastocysts. Prmt5 is also up-regulated in the cytoplasm during the derivation of embryonic stem (ES) cells together with Stat3, where they persist to maintain pluripotency. Prmt5 in association with Mep50 methylates cytosolic histone H2A (H2AR3me2s) to repress differentiation genes in ES cells. Loss of Prmt5 or Mep50 results in derepression of differentiation genes, indicating the significance of the Prmt5/Mep50 complex for pluripotency, which may occur in conjunction with the leukemia inhibitory factor (LIF)/Stat3 pathway.


Assuntos
Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes/citologia , Proteínas Metiltransferases/fisiologia , Animais , Diferenciação Celular/genética , Citoplasma/química , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Metilação , Camundongos , Proteína-Arginina N-Metiltransferases
12.
Nucleic Acids Res ; 37(15): 4965-76, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19528079

RESUMO

Protein arginine methyltransferases (PRMTs) mediate the transfer of methyl groups to arginines in proteins involved in signal transduction, transcriptional regulation and RNA processing. Tumor suppressor p53 coordinates crucial cellular processes, including cell-cycle arrest and DNA repair, in response to stress signals. Post-translational modifications and interactions with co-factors are important to regulate p53 transcriptional activity. To explore whether PRMTs modulate p53 function, we generated multiple cell lines in which PRMT1, CARM1 and PRMT5 are inducibly knocked down. Here, we showed that PRMT5, but not PRMT1 or CARM1, is essential for cell proliferation and PRMT5 deficiency triggers cell-cycle arrest in G1. In addition, PRMT5 is required for p53 expression and induction of p53 targets MDM2 and p21 upon DNA damage. Importantly, we established that PRMT5 knockdown prevents p53 protein synthesis. Furthermore, we found that PRMT5 regulates the expression of translation initiation factor eIF4E and growth suppression mediated upon PRMT5 knockdown is independent of p53 but is dependent on eIF4E. Taken together, we uncovered that arginine methyltransferase PRMT5 is a major pro-survival factor regulating eIF4E expression and p53 translation.


Assuntos
Ciclo Celular , Proteínas Metiltransferases/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Fator de Iniciação 4E em Eucariotos/metabolismo , Fase G1 , Técnicas de Silenciamento de Genes , Humanos , Mutação , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética
13.
Oncogene ; 28(13): 1626-38, 2009 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-19219072

RESUMO

Constitutive nuclear factor (NF)-kappaB activation in haematological malignancies is caused in several cases by loss of function mutations within the coding sequence of NF-kappaB inhibitory molecules such as IkappaBalpha or p100. Hut-78, a truncated form of p100, constitutively generates p52 and contributes to the development of T-cell lymphomas but the molecular mechanism underlying this oncogenic potential remains unclear. We show here that MMP9 gene expression is induced through the alternative NF-kappaB-activating pathway in fibroblasts and also on Hut-78 or p52 overexpression in fibroblasts as well as in lymphoma cells. p52 is critical for Hut-78-mediated MMP9 gene induction as a Hut-78 mutant as well as other truncated NF-kappaB2 proteins that are not processed into p52 failed to induce the expression of this metalloproteinase. Conversely, MMP9 gene expression is impaired in p52-depleted HUT-78 cells. Interestingly, MLL1 and MLL2 H3K4 methyltransferase complexes are tethered by p52 on the MMP9 but not on the IkappaBalpha promoter, and the H3K4 trimethyltransferase activity recruited on the MMP9 promoter is impaired in p52-depleted HUT-78 cells. Moreover, MLL1 and MLL2 are associated with Hut-78 in a native chromatin-enriched extract. Thus, we identified a molecular mechanism by which the recruitment of a H3K4 histone methyltransferase complex on the promoter of a NF-kappaB-dependent gene induces its expression and potentially the invasive potential of lymphoma cells harbouring constitutive activity of the alternative NF-kappaB-activating pathway.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Metaloproteinase 9 da Matriz/biossíntese , Proteína de Leucina Linfoide-Mieloide/metabolismo , Subunidade p52 de NF-kappa B/farmacologia , Proteínas de Neoplasias/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Proteínas de Ligação a DNA/fisiologia , Indução Enzimática/efeitos dos fármacos , Indução Enzimática/fisiologia , Células HeLa , Histona Metiltransferases , Histona-Lisina N-Metiltransferase , Humanos , Lisina/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/fisiologia , Camundongos , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/fisiologia , Proteínas Mutantes/farmacologia , Proteína de Leucina Linfoide-Mieloide/fisiologia , Subunidade p52 de NF-kappa B/química , Células NIH 3T3 , Proteínas de Neoplasias/fisiologia , Proteínas de Fusão Oncogênica/farmacologia , Proteínas Metiltransferases/metabolismo , Proteínas Metiltransferases/fisiologia , Homologia de Sequência de Aminoácidos
14.
Curr Mol Med ; 8(8): 805-15, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19075677

RESUMO

Menin is a tumor suppressor encoded by the MEN1 gene that is mutated in patients with an inherited syndrome, multiple endocrine neoplasia type 1 (MEN1). Loss of menin has potent impact on proliferation of endocrine and non-endocrine cells. However, until recently little has been known as to how menin regulates cell proliferation. Rapid research progress in the past several years suggests that menin represses proliferation of endocrine cells yet promotes proliferation in certain types of leukemia cells via interacting with various transcriptional regulators. Menin interacts with histone H3 methyltransferases such as MLL (mixed lineage leukemia) protein. Increasing evidence has linked the biological function of menin to epigenetic histone modifications, control of the pattern of gene expression, and regulation of cell proliferation in a cell type-specific manner. In light of these recent findings, an emerging model suggests that menin is a crucial regulator of histone modifiers by acting as a scaffold protein to coordinate gene transcription and cell proliferation in a cell context-dependent manner. This recent progress unravels the coordinating role of menin in epigenetics and regulation of cell cycle, providing novel insights into understanding regulation of beta cell functions and diabetes, as well as the development and therapy of endocrine tumors and leukemia.


Assuntos
Proliferação de Células , Proteínas Metiltransferases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Genes Supressores de Tumor , Histona Metiltransferases , Histona-Lisina N-Metiltransferase , Humanos , Leucemia Aguda Bifenotípica/genética , Leucemia Aguda Bifenotípica/patologia , Leucemia Aguda Bifenotípica/fisiopatologia , Modelos Biológicos , Neoplasia Endócrina Múltipla Tipo 1/genética , Neoplasia Endócrina Múltipla Tipo 1/patologia , Neoplasia Endócrina Múltipla Tipo 1/fisiopatologia , Proteína de Leucina Linfoide-Mieloide/genética , Proteína de Leucina Linfoide-Mieloide/fisiologia , Proteínas Proto-Oncogênicas/genética , Translocação Genética
15.
Cell Mol Life Sci ; 65(21): 3329-47, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18818875

RESUMO

Endogenous retrovirus-like elements, or ERVs, are an abundant component of all eukaryotic genomes. Their transcriptional and retrotranspositional activities have great potential for deleterious effects on gene expression. Consequences of such activity may include germline mutagenesis and cancerous transformation. As a result, mammalian genomes have evolved means of counteracting ERV transcription and mobilization. In this review, we discuss epigenetic mechanisms of ERV and LTR retrotransposon control during mouse development, focusing on involvement of DNA methylation, histone modifications, small RNAs and their interaction with one another. We also address relevance of research performed in the mouse system to human and challenges associated with studying repetitive families. (Part of a multi-author review).


Assuntos
Retrovirus Endógenos/genética , Epigênese Genética/fisiologia , Regulação Viral da Expressão Gênica , Inativação Gênica/fisiologia , Interações Hospedeiro-Patógeno/genética , Camundongos/virologia , Retroelementos/genética , Animais , Blastocisto , Transformação Celular Viral/genética , DNA (Citosina-5-)-Metiltransferases/fisiologia , Desenvolvimento Embrionário/genética , Epigênese Genética/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Células Germinativas/virologia , Histona Metiltransferases , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Humanos , Masculino , Metilação , Camundongos/embriologia , Proteínas Metiltransferases/fisiologia , Processamento de Proteína Pós-Traducional , Peixe-Zebra/embriologia , Peixe-Zebra/genética
16.
EMBO Rep ; 9(5): 452-8, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18404153

RESUMO

Protein arginine methyltransferase 5 (PRMT5) targets nuclear and cytoplasmic proteins. Here, we identified a nuclear protein, called cooperator of PRMT5 (COPR5), involved in the nuclear functions of PRMT5. COPR5 tightly binds to PRMT5, both in vitro and in living cells, but not to other members of the PRMT family. PRMT5 bound to COPR5 methylates histone H4 (R3) preferentially when compared with histone H3 (R8), suggesting that COPR5 modulates the substrate specificity of nuclear PRMT5-containing complexes, at least towards histones. Markedly, recombinant COPR5 binds to the amino terminus of histone H4 and is required to recruit PRMT5 to reconstituted nucleosomes in vitro. Consistently, COPR5 depletion in cells strongly reduces PRMT5 recruitment on chromatin at the PRMT5 target gene cyclin E1 (CCNE1) in vivo. Moreover, both COPR5 depletion and overexpression affect CCNE1 promoter expression. We propose that COPR5 is an important chromatin adaptor for PRMT5 to function on a subset of its target genes.


Assuntos
Arginina/metabolismo , Proteínas de Transporte/fisiologia , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Metiltransferases/fisiologia , Sequência de Aminoácidos , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/química , Núcleo Celular/metabolismo , Cromatina/metabolismo , Metilação de DNA , Genes Reporter , Glutationa Transferase/metabolismo , Humanos , Luciferases/metabolismo , Dados de Sequência Molecular , Proteínas Nucleares/fisiologia , Ligação Proteica , Proteínas Metiltransferases/metabolismo , Estrutura Secundária de Proteína , Proteínas Recombinantes/metabolismo
17.
Mol Cell Biol ; 27(1): 384-94, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17043109

RESUMO

Skeletal muscle differentiation requires the coordinated activity of transcription factors, histone modifying enzymes, and ATP-dependent chromatin remodeling enzymes. The type II protein arginine methyltransferase Prmt5 symmetrically dimethylates histones H3 and H4 and numerous nonchromatin proteins, and prior work has implicated Prmt5 in transcriptional repression. Here we demonstrate that MyoD-induced muscle differentiation requires Prmt5. One of the first genes activated during differentiation encodes the myogenic regulator myogenin. Prmt5 and dimethylated H3R8 (histone 3 arginine 8) are localized at the myogenin promoter in differentiating cells. Modification of H3R8 required Prmt5, and reduction of Prmt5 resulted in the abrogation of promoter binding by the Brg1 ATPase-associated with the SWI/SNF chromatin remodeling enzymes and all subsequent events associated with gene activation, including increases in chromatin accessibility and stable binding by MyoD. Prmt5 and dimethylated H3R8 were also associated with the myogenin promoter in activated satellite cells isolated from muscle tissue, further demonstrating the physiological relevance of these observations. The data indicate that Prmt5 facilitates myogenesis because it is required for Brg1-dependent chromatin remodeling and gene activation at a locus essential for differentiation. We therefore conclude that a histone modifying enzyme is necessary to permit an ATP-dependent chromatin remodeling enzyme to function.


Assuntos
Trifosfato de Adenosina/química , Cromatina/química , Músculos/metabolismo , Proteínas Metiltransferases/fisiologia , Animais , Diferenciação Celular , Separação Celular , Cromatina/metabolismo , DNA Helicases/metabolismo , Metilação de DNA , Enzimas de Restrição do DNA/metabolismo , Citometria de Fluxo , Camundongos , Músculo Esquelético/metabolismo , Células NIH 3T3 , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteínas Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
18.
Mol Microbiol ; 61(6): 1636-45, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16968230

RESUMO

In contrast to most primary metabolism genes, the genes involved in secondary metabolism and certain nutrient utilization pathways are clustered in fungi. Recently a nuclear protein, LaeA, was found to be required for the transcription of several secondary metabolite gene clusters in Aspergillus nidulans. Here we show that LaeA regulation does not extend to nutrient utilization or the spoC1 sporulation clusters. One of the secondary metabolite clusters regulated by LaeA contains the positive regulatory (i.e. aflR) and biosynthetic genes required for biosynthesis of sterigmatocystin (ST), a carcinogenic toxin. Analysis of ST gene cluster expression indicates LaeA regulation of the cluster is location specific as transcription of genes bordering the ST cluster are unaffected in a DeltalaeA mutant and placement of a primary metabolic gene, argB, in the ST cluster resulted in argB silencing in the DeltalaeA background. ST cluster gene expression was remediated when an additional copy of aflR was placed outside of the cluster but not when placed in the cluster. Site-specific mutation of an s-adenosyl methionine (AdoMet) binding site in LaeA generated a DeltalaeA phenotype suggesting the protein to be a methyltransferase.


Assuntos
Aspergillus nidulans/genética , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Inativação Gênica , Proteínas Metiltransferases/fisiologia , Esterigmatocistina/metabolismo , Fatores de Transcrição/fisiologia , Aspergillus nidulans/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Proteínas Fúngicas/fisiologia , Ordem dos Genes , Metilação , Família Multigênica/genética , Proteínas Metiltransferases/genética , Processamento de Proteína Pós-Traducional , Fatores de Transcrição/genética
19.
J Biol Chem ; 281(28): 19489-500, 2006 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-16682412

RESUMO

DNA CpG methylation can cooperate with histone H3 lysine 9 (H3-K9) methylation in heterochromatin formation and gene silencing. Trimethylation of H3-K9 by the recently identified euchromatic histone methyltransferase SETDB1/ESET may be responsible for transcriptional repression of certain promoters. Here, we show that SETDB1 associates with endogenous DNA methyltransferase activity. SETDB1 interacts with the de novo DNA methyltransferases DNMT3A and DNMT3B but not with the maintenance methyltransferase DNMT1. The interaction of SETDB1 with DNMT3A was further characterized and confirmed by in vivo and in vitro interaction studies. A direct interaction of the two proteins occurs through the N terminus of SETDB1 and the plant homeodomain of DNMT3A. Co-expression of SETDB1 and DNMT3A was essential for repression of reporter gene expression in a Gal4-based tethering assay and resulted in their recruitment to the artificial promoter. We further demonstrate that the CpG-methylated promoters of the endogenous p53BP2 gene in HeLa cells and the RASSF1A gene in MDA-MB-231 cells are simultaneously occupied by both SETDB1 and DNMT3A proteins, which provides evidence for SETDB1 being at least partly responsible for H3-K9 trimethylation at the promoter of RASSF1A, a gene frequently silenced in human cancers. In summary, our data demonstrate the direct physical interaction and functional connection between the H3-K9 trimethylase SETDB1 and the DNA methyltransferase DNMT3A and thus contribute to a better understanding of the complexity of the self-reinforcing heterochromatin machinery operating at silenced promoters.


Assuntos
DNA (Citosina-5-)-Metiltransferases/fisiologia , Neoplasias/metabolismo , Regiões Promotoras Genéticas , Proteínas Metiltransferases/fisiologia , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Ilhas de CpG , Metilação de DNA , DNA Metiltransferase 3A , Inativação Gênica , Células HeLa , Heterocromatina/metabolismo , Histona-Lisina N-Metiltransferase , Humanos , Proteínas Supressoras de Tumor/metabolismo
20.
J Biol Chem ; 280(19): 18790-6, 2005 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-15647276

RESUMO

We have previously demonstrated that both isoprenylcysteine carboxylmethyltransferase (ICMT) and one of its substrates, the RhoGTPase Rac1, are critical for the tumor necrosis factor alpha (TNF alpha) stimulation of vascular cell adhesion molecule-1 expression in endothelial cells (EC). Here, we have shown that ICMT regulates TNF alpha stimulation of Rac1 activity. TNF alpha stimulation of EC increased the membrane association of Rac1, an event that is essential for Rac1 activity. ICMT inhibitor N-acetyl-S-farnesyl-L-cysteine (AFC) blocked the accumulation of Rac1 into the membrane both in resting and TNF alpha-stimulated conditions. Similarly, the membrane-associated Rac1 was lower in Icmt-deficient versus wild-type mouse embryonic fibroblasts (MEFs). TNF alpha also increased the level of GTP-Rac1, the active form of Rac1, in EC. AFC completely suppressed the TNF alpha stimulation of increase in GTP-Rac1 levels. Confocal microscopy revealed resting EC Rac1 was present in the plasma membrane and also in the perinuclear region. AFC mislocalized Rac1, both from the plasma membrane and the perinuclear region. Mislocalization of Rac1 was also observed in Icmt-deficient versus wild-type MEFs. To determine the consequences of ICMT inhibition, we investigated the effect of AFC on p38 mitogen-activated protein (MAP) kinase phosphorylation, which is downstream of Rac1. AFC inhibited the TNF alpha stimulation of p38 MAP kinase phosphorylation in EC. TNF alpha stimulation of p38 MAP kinase phosphorylation was also significantly attenuated in Icmt-deficient versus wild-type MEFs. To understand the mechanism of inhibition of Rac1 activity, we examined the effect of ICMT inhibition on the interaction of Rac1 with its inhibitor, Rho guanine nucleotide dissociation inhibitor (RhoGDI). The association of Rac1 with its inhibitor RhoGDI was dramatically increased in the Icmt-deficient versus wild-type MEFs both in resting as well as in TNF alpha-stimulated conditions, suggesting that RhoGDI was involved in inhibiting Rac1 activity under the conditions of ICMT inhibition. These results suggest that ICMT regulates Rac1 activity by controlling the interaction of Rac1 with RhoGDI. We hypothesize that ICMT regulates the release of Rac1 from RhoGDI.


Assuntos
Regulação Enzimológica da Expressão Gênica , Proteínas Metiltransferases/química , Fator de Necrose Tumoral alfa/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Aorta/patologia , Western Blotting , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Detergentes/farmacologia , Eletroforese em Gel Bidimensional , Células Endoteliais/metabolismo , Endotélio Vascular/patologia , Fibroblastos/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Metilação , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Fosforilação , Proteínas Metiltransferases/fisiologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA