Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Life Sci Alliance ; 4(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33579760

RESUMO

Isoprenylcysteine carboxyl methyltransferase (ICMT) is the third of three enzymes that sequentially modify the C-terminus of CaaX proteins, including RAS. Although all four RAS proteins are substrates for ICMT, each traffics to membranes differently by virtue of their hypervariable regions that are differentially palmitoylated. We found that among RAS proteins, NRAS was unique in requiring ICMT for delivery to the PM, a consequence of having only a single palmitoylation site as its secondary affinity module. Although not absolutely required for palmitoylation, acylation was diminished in the absence of ICMT. Photoactivation and FRAP of GFP-NRAS revealed increase flux at the Golgi, independent of palmitoylation, in the absence of ICMT. Association of NRAS with the prenyl-protein chaperone PDE6δ also required ICMT and promoted anterograde trafficking from the Golgi. We conclude that carboxyl methylation of NRAS is required for efficient palmitoylation, PDE6δ binding, and homeostatic flux through the Golgi, processes that direct delivery to the plasma membrane.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Metiltransferases/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Movimento Celular/fisiologia , GTP Fosfo-Hidrolases/fisiologia , Humanos , Lipoilação/fisiologia , Proteínas de Membrana/fisiologia , Proteínas Metiltransferases/fisiologia , Transporte Proteico/fisiologia , Proteínas ras
2.
Nat Commun ; 8(1): 2057, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29234025

RESUMO

SETDB1 is an essential H3K9 methyltransferase involved in silencing of retroviruses and gene regulation. We show here that its triple Tudor domain (3TD) specifically binds to doubly modified histone H3 containing K14 acetylation and K9 methylation. Crystal structures of 3TD in complex with H3K14ac/K9me peptides reveal that peptide binding and K14ac recognition occurs at the interface between Tudor domains (TD) TD2 and TD3. Structural and biochemical data demonstrate a pocket switch mechanism in histone code reading, because K9me1 or K9me2 is preferentially recognized by the aromatic cage of TD3, while K9me3 selectively binds to TD2. Mutations in the K14ac/K9me binding sites change the sub-nuclear localization of 3TD. ChIP-seq analyses show that SETDB1 is enriched at H3K9me3 regions and K9me3/K14ac is enriched at SETDB1 binding sites overlapping with LINE elements, suggesting that recruitment of the SETDB1 complex to K14ac/K9me regions has a role in silencing of active genomic regions.


Assuntos
Histonas/metabolismo , Elementos Nucleotídeos Longos e Dispersos/fisiologia , Proteínas Metiltransferases/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Domínio Tudor/fisiologia , Acetilação , Animais , Sítios de Ligação/fisiologia , Cristalografia por Raios X , Células HEK293 , Histona-Lisina N-Metiltransferase , Histonas/genética , Humanos , Metilação , Camundongos , Células-Tronco Embrionárias Murinas , Ligação Proteica/fisiologia , Proteínas Metiltransferases/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
3.
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
4.
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
5.
Biol Reprod ; 92(4): 104, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25810472

RESUMO

In mammals, germ cells undergo massive epigenetic remodeling during fetal development. However, the physiological functions of epigenetic modification in germ cell development remain unclear. In this study, we found that protein arginine methyltransferase 5 (Prmt5) was abundantly expressed in the germ cells of both male and female gonads. Deletion of Prmt5 by crossing with Tnap-Cre mice resulted in germ cell depletion in adult mice. Germ cell loss was first observed between Embryonic Days 12.5 and 13.5 (E12.5 and E13.5), and very few of these cells remained at birth. Oct4, Sox2, and Nanog were abundantly expressed in Prmt5-deficient germ cells at E13.5 and E15.5, whereas the expression of these genes was dramatically decreased in control germ cells. Interestingly, the expression of meiosis-associated genes was virtually absent in Prmt5-deficient female germ cells at E13.5, whereas the expression of other germ cell-specific genes was not changed. Further study revealed that H4R3me2s was completely absent after Prmt5 inactivation, whereas the level of H3R2me2s was not changed in Prmt5-deficient germ cells. Collectively, this study demonstrated that Prmt5 plays critical roles in germ cell development that are required for germ cell survival during embryonic stages.


Assuntos
Desenvolvimento Embrionário/fisiologia , Células Germinativas/fisiologia , Proteínas Metiltransferases/fisiologia , Animais , Sobrevivência Celular/fisiologia , Feminino , Histonas/metabolismo , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Imuno-Histoquímica , Masculino , Meiose/fisiologia , Camundongos , Camundongos Transgênicos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/biossíntese , Fator 3 de Transcrição de Octâmero/genética , Gravidez , Proteína-Arginina N-Metiltransferases , Fatores de Transcrição SOXB1/biossíntese , Fatores de Transcrição SOXB1/genética , Regulação para Cima/fisiologia
7.
Mol Cell ; 56(4): 564-79, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25457166

RESUMO

Primordial germ cells (PGCs) and preimplantation embryos undergo epigenetic reprogramming, which includes comprehensive DNA demethylation. We found that PRMT5, an arginine methyltransferase, translocates from the cytoplasm to the nucleus during this process. Here we show that conditional loss of PRMT5 in early PGCs causes complete male and female sterility, preceded by the upregulation of LINE1 and IAP transposons as well as activation of a DNA damage response. Similarly, loss of maternal-zygotic PRMT5 also leads to IAP upregulation. PRMT5 is necessary for the repressive H2A/H4R3me2s chromatin modification on LINE1 and IAP transposons in PGCs, directly implicating this modification in transposon silencing during DNA hypomethylation. PRMT5 translocates back to the cytoplasm subsequently, to participate in the previously described PIWI-interacting RNA (piRNA) pathway that promotes transposon silencing via de novo DNA remethylation. Thus, PRMT5 is directly involved in genome defense during preimplantation development and in PGCs at the time of global DNA demethylation.


Assuntos
Blastocisto/enzimologia , Metilação de DNA , Instabilidade Genômica , Óvulo/enzimologia , Proteínas Metiltransferases/fisiologia , Espermatozoides/enzimologia , Animais , Apoptose , Blastocisto/citologia , Células Cultivadas , Dano ao DNA , Elementos de DNA Transponíveis , Desenvolvimento Embrionário , Células-Tronco Embrionárias/enzimologia , Feminino , Histonas/metabolismo , Masculino , Camundongos Transgênicos , Processamento de Proteína Pós-Traducional , Proteína-Arginina N-Metiltransferases
8.
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
9.
J Biol Chem ; 289(44): 30499-30510, 2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25231979

RESUMO

The components of the cellular protein translation machinery, such as ribosomal proteins and translation factors, are subject to numerous post-translational modifications. In particular, this group of proteins is frequently methylated. However, for the majority of these methylations, the responsible methyltransferases (MTases) remain unknown. The human FAM86A (family with sequence similarity 86) protein belongs to a recently identified family of protein MTases, and we here show that FAM86A catalyzes the trimethylation of eukaryotic elongation factor 2 (eEF2) on Lys-525. Moreover, we demonstrate that the Saccharomyces cerevisiae MTase Yjr129c, which displays sequence homology to FAM86A, is a functional FAM86A orthologue, modifying the corresponding residue (Lys-509) in yeast eEF2, both in vitro and in vivo. Finally, Yjr129c-deficient yeast cells displayed phenotypes related to eEF2 function (i.e. increased frameshifting during protein translation and hypersensitivity toward the eEF2-specific drug sordarin). In summary, the present study establishes the function of the previously uncharacterized MTases FAM86A and Yjr129c, demonstrating that these enzymes introduce a functionally important lysine methylation in eEF2. Based on the previous naming of similar enzymes, we have redubbed FAM86A and Yjr129c as eEF2-KMT and Efm3, respectively.


Assuntos
Metiltransferases/genética , Fator 2 de Elongação de Peptídeos/metabolismo , Proteínas Metiltransferases/fisiologia , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/enzimologia , Sequência de Aminoácidos , Animais , Sequência Conservada , Células HEK293 , Humanos , Metilação , Metiltransferases/metabolismo , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional , Coelhos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
10.
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
11.
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
12.
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
13.
PLoS One ; 7(10): e48152, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23133559

RESUMO

Circadian clocks are the endogenous oscillators that regulate rhythmic physiological and behavioral changes to correspond to daily light-dark cycles. Molecular dissections have revealed that transcriptional feedback loops of the circadian clock genes drive the molecular oscillation, in which PER/CRY complexes inhibit the transcriptional activity of the CLOCK/BMAL1 heterodimer to constitute a negative feedback loop. In this study, we identified the type II protein arginine methyltransferase 5 (PRMT5) as an interacting molecule of CRY1. Although the Prmt5 gene was constitutively expressed, increased interaction of PRMT5 with CRY1 was observed when the Per1 gene was repressed both in synchronized mouse liver and NIH3T3 cells. Moreover, rhythmic recruitment of PRMT5 and CRY1 to the Per1 gene promoter was found to be associated with an increased level of histone H4R3 dimethylation and Per1 gene repression. Consistently, decreased histone H4R3 dimethylation and altered rhythmic Per1 gene expression were observed in Prmt5-depleted cells. Taken together, these findings provide an insight into the link between histone arginine methylation by PRMT5 and transcriptional regulation of the circadian Per1 gene.


Assuntos
Criptocromos/biossíntese , Regulação Enzimológica da Expressão Gênica , Proteínas Circadianas Period/biossíntese , Proteínas Metiltransferases/fisiologia , Proteína-Arginina N-Metiltransferases/fisiologia , Animais , Especificidade de Anticorpos , Arginina/química , Células HEK293 , Histonas/química , Histonas/metabolismo , Humanos , Metilação , Camundongos , Modelos Genéticos , Células NIH 3T3 , Regiões Promotoras Genéticas , Transcrição Gênica
14.
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
15.
Biochim Biophys Acta ; 1819(7): 727-32, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22240386

RESUMO

Histone methylation is implicated in both gene activation and repression, depending on the specific lysine residue that gets methylated. Recent years have witnessed an explosive expansion of the list of remarkably site-specific histone methyltransferases and demethylases, which greatly facilitates the study on the biological functions of histone methylation in gene expression and cell differentiation in mammalian cells. Adipogenesis represents an excellent model system to understand transcriptional and epigenetic regulation of gene expression and cell differentiation. While transcriptional regulation of adipogenesis has been extensively studied, the roles of epigenetic mechanisms in particular histone methylation in regulation of adipogenesis have just begun to be understood. This review will summarize the recent progress on epigenetic regulation of adipogenesis by histone methylation, with a focus on histone H3K4 and H3K27. The available evidence suggests that site-specific histone methylations play critical roles in adipogenesis and control the expression of both positive and negative master regulators of adipogenesis. This article is part of a Special Issue entitled: Chromatin in time and space.


Assuntos
Adipogenia/genética , Epigênese Genética , Histonas/metabolismo , Sequência de Aminoácidos , Animais , Histona Desmetilases/metabolismo , Histona Desmetilases/fisiologia , Histonas/química , Humanos , Metilação , Dados de Sequência Molecular , Proteínas Metiltransferases/metabolismo , Proteínas Metiltransferases/fisiologia , Processamento de Proteína Pós-Traducional
16.
Mol Cell ; 45(2): 185-95, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22169276

RESUMO

The repression of transcription, through the concerted actions of tissue specific DNA binding proteins, Polycomb repressor complexes, and DNA methylation, is essential for maintaining stem cell pluripotency and for cell fate specification in development. In this report, we show that recruitment of the co-repressor protein Grg4 to a Pax DNA-binding site displaces the adaptor protein PTIP and a histone H3K4me complex. Grg4 recruits the arginine methyltransferase PRMT5 to chromatin resulting in symmetric H4R3 dimethylation. PRMT5 is essential for recruiting Polycomb proteins, in a Pax2/Grg4 dependent manner, which results in H3K27 methylation. These data define the early epigenetic events in response to Pax/Grg mediated gene repression and demonstrate that a single DNA binding protein can recruit either an activator or a repressor complex depending on the availability of Grg4. These data suggest a model for understanding the initiation of Groucho/Grg/TLE mediated gene silencing.


Assuntos
Epigênese Genética , Histonas/metabolismo , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Proteínas de Transporte/metabolismo , Cromatina/metabolismo , Proteínas de Ligação a DNA , Inativação Gênica , Células HEK293 , Humanos , Metilação , Modelos Genéticos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas do Grupo Polycomb , Proteínas Metiltransferases/metabolismo , Proteínas Metiltransferases/fisiologia , Transporte Proteico , Proteína-Arginina N-Metiltransferases , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
17.
Biochem J ; 441(3): 803-12, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22004688

RESUMO

PABP1 [poly(A)-binding protein 1] is a central regulator of mRNA translation and stability and is required for miRNA (microRNA)-mediated regulation and nonsense-mediated decay. Numerous protein, as well as RNA, interactions underlie its multi-functional nature; however, it is unclear how its different activities are co-ordinated, since many partners interact via overlapping binding sites. In the present study, we show that human PABP1 is subject to elaborate post-translational modification, identifying 14 modifications located throughout the functional domains, all but one of which are conserved in mouse. Intriguingly, PABP1 contains glutamate and aspartate methylations, modifications of unknown function in eukaryotes, as well as lysine and arginine methylations, and lysine acetylations. The latter dramatically alter the pI of PABP1, an effect also observed during the cell cycle, suggesting that different biological processes/stimuli can regulate its modification status, although PABP1 also probably exists in differentially modified subpopulations within cells. Two lysine residues were differentially acetylated or methylated, revealing that PABP1 may be the first example of a cytoplasmic protein utilizing a 'methylation/acetylation switch'. Modelling using available structures implicates these modifications in regulating interactions with individual PAM2 (PABP-interacting motif 2)-containing proteins, suggesting a direct link between PABP1 modification status and the formation of distinct mRNP (messenger ribonucleoprotein) complexes that regulate mRNA fate in the cytoplasm.


Assuntos
Proteína I de Ligação a Poli(A)/química , Proteína I de Ligação a Poli(A)/metabolismo , Proteína I de Ligação a Poli(A)/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Animais , Arginina/metabolismo , Células Cultivadas , Células HeLa , Humanos , Cinética , Metilação , Camundongos , Modelos Moleculares , Proteína I de Ligação a Poli(A)/genética , Proteínas Metiltransferases/metabolismo , Proteínas Metiltransferases/fisiologia , Processamento de Proteína Pós-Traducional/genética , Relação Estrutura-Atividade , Distribuição Tecidual
18.
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
19.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA