Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Immunol ; 17(2): 140-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26657003

RESUMO

Innate sensing of pathogens initiates inflammatory cytokine responses that need to be tightly controlled. We found here that after engagement of Toll-like receptors (TLRs) in myeloid cells, deficient sumoylation caused increased secretion of transcription factor NF-κB-dependent inflammatory cytokines and a massive type I interferon signature. In mice, diminished sumoylation conferred susceptibility to endotoxin shock and resistance to viral infection. Overproduction of several NF-κB-dependent inflammatory cytokines required expression of the type I interferon receptor, which identified type I interferon as a central sumoylation-controlled hub for inflammation. Mechanistically, the small ubiquitin-like modifier SUMO operated from a distal enhancer of the gene encoding interferon-ß (Ifnb1) to silence both basal and stimulus-induced activity of the Ifnb1 promoter. Therefore, sumoylation restrained inflammation by silencing Ifnb1 expression and by strictly suppressing an unanticipated priming by type I interferons of the TLR-induced production of inflammatory cytokines.


Assuntos
Resistência à Doença , Regulação da Expressão Gênica , Imunidade Inata , Imunomodulação , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Sumoilação , Animais , Cromatina/genética , Cromatina/metabolismo , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Elementos Facilitadores Genéticos , Perfilação da Expressão Gênica , Loci Gênicos , Inflamação/virologia , Mediadores da Inflamação/metabolismo , Interferon beta/metabolismo , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Knockout , Ligação Proteica , Receptor de Interferon alfa e beta/metabolismo , Elementos Reguladores de Transcrição , Proteína SUMO-1/metabolismo , Choque Séptico/genética , Choque Séptico/imunologia , Choque Séptico/metabolismo , Transdução de Sinais , Sumoilação/genética , Sumoilação/imunologia , Receptores Toll-Like/metabolismo
2.
Genome Res ; 23(10): 1563-79, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23893515

RESUMO

Despite numerous studies on specific sumoylated transcriptional regulators, the global role of SUMO on chromatin in relation to transcription regulation remains largely unknown. Here, we determined the genome-wide localization of SUMO1 and SUMO2/3, as well as of UBC9 (encoded by UBE2I) and PIASY (encoded by PIAS4), two markers for active sumoylation, along with Pol II and histone marks in proliferating versus senescent human fibroblasts together with gene expression profiling. We found that, whereas SUMO alone is widely distributed over the genome with strong association at active promoters, active sumoylation occurs most prominently at promoters of histone and protein biogenesis genes, as well as Pol I rRNAs and Pol III tRNAs. Remarkably, these four classes of genes are up-regulated by inhibition of sumoylation, indicating that SUMO normally acts to restrain their expression. In line with this finding, sumoylation-deficient cells show an increase in both cell size and global protein levels. Strikingly, we found that in senescent cells, the SUMO machinery is selectively retained at histone and tRNA gene clusters, whereas it is massively released from all other unique chromatin regions. These data, which reveal the highly dynamic nature of the SUMO landscape, suggest that maintenance of a repressive environment at histone and tRNA loci is a hallmark of the senescent state. The approach taken in our study thus permitted the identification of a common biological output and uncovered hitherto unknown functions for active sumoylation at chromatin as a key mechanism that, in dynamically marking chromatin by a simple modifier, orchestrates concerted transcriptional regulation of a network of genes essential for cell growth and proliferation.


Assuntos
Proliferação de Células , Cromatina/genética , Cromatina/metabolismo , Regulação da Expressão Gênica , Genes Essenciais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Animais , Ciclo Celular , Linhagem Celular , Senescência Celular , Perfilação da Expressão Gênica , Histonas/genética , Histonas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , RNA de Transferência/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Sumoilação , Transcrição Gênica , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo
3.
EMBO J ; 28(22): 3534-48, 2009 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-19779455

RESUMO

Heat shock and other environmental stresses rapidly induce transcriptional responses subject to regulation by a variety of post-translational modifications. Among these, poly(ADP-ribosyl)ation and sumoylation have received growing attention. Here we show that the SUMO E3 ligase PIASy interacts with the poly(ADP-ribose) polymerase PARP-1, and that PIASy mediates heat shock-induced poly-sumoylation of PARP-1. Furthermore, PIASy, and hence sumoylation, appears indispensable for full activation of the inducible HSP70.1 gene. Chromatin immunoprecipitation experiments show that PIASy, SUMO and the SUMO-conjugating enzyme Ubc9 are rapidly recruited to the HSP70.1 promoter upon heat shock, and that they are subsequently released with kinetics similar to PARP-1. Finally, we provide evidence that the SUMO-targeted ubiquitin ligase RNF4 mediates heat-shock-inducible ubiquitination of PARP-1, regulates the stability of PARP-1, and, like PIASy, is a positive regulator of HSP70.1 gene activity. These results, thus, point to a novel mechanism for regulating PARP-1 transcription function, and suggest crosstalk between sumoylation and RNF4-mediated ubiquitination in regulating gene expression in response to heat shock.


Assuntos
Resposta ao Choque Térmico/fisiologia , Poli(ADP-Ribose) Polimerases/metabolismo , Poli(ADP-Ribose) Polimerases/fisiologia , Proteína SUMO-1/metabolismo , Ativação Transcricional , Animais , Células Cultivadas , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Células Jurkat , Camundongos , Modelos Biológicos , Poli(ADP-Ribose) Polimerase-1 , Proteínas de Ligação a Poli-ADP-Ribose , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas Inibidoras de STAT Ativados/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Spodoptera , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/fisiologia
4.
Am J Hum Genet ; 87(4): 505-12, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20887963

RESUMO

One in seven couples worldwide are infertile, and male factor infertility accounts for approximately 30%-50% of these cases. Although many genes are known to be essential for gametogenesis, there are surprisingly few monogenic mutations that have been conclusively demonstrated to cause human spermatogenic failure. A nuclear receptor, NR5A1 (also called steroidogenic factor 1), is a key transcriptional regulator of genes involved in the hypothalamic-pituitary-steroidogenic axis, and it is expressed in the steroidogenic tissue of the developing and adult human gonad. Mutations of NR5A1 have been reported in 46,XY disorders of sex development and in 46,XX primary ovarian insufficiency. To test the hypothesis that mutations in NR5A1 cause male infertility, we sequenced NR5A1 in 315 men with idiopathic spermatogenic failure. We identified seven men with severe spermatogenic failure who carried missense mutations in NR5A1. Functional studies indicated that these mutations impaired NR5A1 transactivational activity. We did not observe these mutations in more than 4000 control alleles, including the entire coding sequence of 359 normospermic men and 370 fertile male controls. NR5A1 mutations are found in approximately 4% of men with otherwise unexplained severe spermatogenic failure.


Assuntos
Infertilidade Masculina/genética , Espermatogênese/genética , Fator Esteroidogênico 1/genética , Ativação Transcricional/genética , Sequência de Aminoácidos , Sequência de Bases , França , Humanos , Masculino , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto/genética , Análise de Sequência de DNA
5.
Proc Natl Acad Sci U S A ; 106(33): 13826-31, 2009 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-19666599

RESUMO

Gene silencing via heterochromatin formation plays a major role in cell differentiation and maintenance of homeostasis. Here we report the identification and characterization of a novel heterochromatinization factor in vertebrates, bromo adjacent homology domain-containing protein 1 (BAHD1). This nuclear protein interacts with HP1, MBD1, HDAC5, and several transcription factors. Through electron and immunofluorescence microscopy studies, we show that BAHD1 overexpression directs HP1 to specific nuclear sites and promotes the formation of large heterochromatic domains, which lack acetyl histone H4 and are enriched in H3 trimethylated at lysine 27 (H3K27me3). Furthermore, ectopically expressed BAHD1 colocalizes with the heterochromatic inactive X chromosome (Xi). The BAH domain is required for BAHD1 colocalization with H3K27me3, but not with the Xi chromosome. As highlighted by whole genome microarray analysis of BAHD1 knockdown cells, BAHD1 represses several proliferation and survival genes, in particular the insulin-like growth factor II gene (IGF2). When overexpressed, BAHD1 specifically binds the CpG-rich P3 promoter of IGF2, which increases MBD1 and HDAC5 targeting at this locus. This region contains DNA-binding sequences for the transcription factor SP1, with which BAHD1 coimmunoprecipitates. Collectively, these findings provide evidence that BAHD1 acts as a silencer by recruiting at specific promoters a set of proteins that coordinate heterochromatin assembly.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Inativação Gênica , Heterocromatina/química , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Cromatina/química , Mapeamento Cromossômico , Ilhas de CpG , Heterocromatina/metabolismo , Histonas/química , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Lisina/química , Microscopia de Fluorescência/métodos , Modelos Genéticos , Ligação Proteica , Estrutura Terciária de Proteína , Transcrição Gênica
6.
Mol Cell Biol ; 27(7): 2661-75, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17283066

RESUMO

Tumor suppressor HIC1 (hypermethylated in cancer 1) is a gene that is essential for mammalian development, epigenetically silenced in many human tumors, and involved in a complex pathway regulating P53 tumor suppression activity. HIC1 encodes a sequence-specific transcriptional repressor containing five Krüppel-like C(2)H(2) zinc fingers and an N-terminal BTB/POZ repression domain. Here, we show that endogenous HIC1 is SUMOylated in vivo on a phylogenetically conserved lysine, K314, located in the central region which is a second repression domain. K314R mutation does not influence HIC1 subnuclear localization but significantly reduces its transcriptional repression potential, as does the mutation of the other conserved residue in the psiKXE consensus, E316A, or the overexpression of the deSUMOylase SSP3/SENP2. Furthermore, HIC1 is acetylated in vitro by P300/CBP. Strikingly, the K314R mutant is less acetylated than wild-type HIC1, suggesting that this lysine is a target for both SUMOylation and acetylation. We further show that HIC1 transcriptional repression activity is positively controlled by two types of deacetylases, SIRT1 and HDAC4, which increase the deacetylation and SUMOylation, respectively, of K314. Knockdown of endogenous SIRT1 by the transfection of short interfering RNA causes a significant loss of HIC1 SUMOylation. Thus, this dual-deacetylase complex induces either a phosphorylation-dependent acetylation-SUMOylation switch through a psiKXEXXSP motif, as previously shown for MEF2, or a phosphorylation-independent switch through a psiKXEP motif, as shown here for HIC1, since P317A mutation severely impairs HIC1 acetylation. Finally, our results demonstrate that HIC1 is a target of the class III deacetylase SIRT1 and identify a new posttranslational modification step in the P53-HIC1-SIRT1 regulatory loop.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteína SUMO-1/metabolismo , Sirtuínas/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Acetilação , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Sequência Conservada , Proteínas de Ligação a DNA/genética , Histona Desacetilases/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like , Lisina/metabolismo , Dados de Sequência Molecular , Mutação , Fosforilação , Filogenia , RNA Interferente Pequeno/genética , Sirtuína 1 , Sirtuínas/genética , Fatores de Transcrição/genética , Transcrição Gênica
7.
Oncogene ; 39(43): 6692-6703, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32948837

RESUMO

Sumoylation is an essential posttranslational modification in eukaryotes that has emerged as an important pathway in oncogenic processes. Most human cancers display hyperactivated sumoylation and many cancer cells are remarkably sensitive to its inhibition, thus supporting application of chemical sumoylation inhibitors in cancer treatment. Here we show, first, that transformed embryonic fibroblasts derived from mice haploinsufficient for Ubc9, the essential and unique gene encoding the SUMO E2 conjugating enzyme, exhibit enhanced proliferation and transformed phenotypes in vitro and as xenografts ex vivo. To then evaluate the possible impact of loss of one Ubc9 allele in vivo, we used a mouse model of intestinal tumorigenesis. We crossed Ubc9+/- mice with mice harboring a conditional ablation of Apc either all along the crypt-villus axis or only in Lgr5+ crypt-based columnar (CBC) cells, the cell compartment that includes the intestinal stem cells proposed as cells-of-origin of intestinal cancer. While Ubc9+/- mice display no overt phenotypes and no globally visible hyposumoylation in cells of the small intestine, we found, strikingly, that, upon loss of Apc in both models, Ubc9+/- mice develop more (>2-fold) intestinal adenomas and show significantly shortened survival. This is accompanied by reduced global sumoylation levels in the polyps, indicating that Ubc9 levels become critical upon oncogenic stress. Moreover, we found that, in normal conditions, Ubc9+/- mice show a moderate but robust (15%) increase in the number of Lgr5+ CBC cells when compared to their wild-type littermates, and further, that these cells display higher degree of stemness and cancer-related and inflammatory gene expression signatures that, altogether, may contribute to enhanced intestinal tumorigenesis. The phenotypes of Ubc9 haploinsufficiency discovered here indicate an unanticipated tumor-suppressive role of sumoylation, one that may have important implications for optimal use of sumoylation inhibitors in the clinic.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Transformação Celular Neoplásica/genética , Neoplasias Intestinais/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Embrião de Mamíferos , Fibroblastos , Haploinsuficiência , Humanos , Mucosa Intestinal/patologia , Neoplasias Intestinais/patologia , Camundongos , Camundongos Transgênicos , Cultura Primária de Células , Transdução de Sinais/genética , Sumoilação/genética , Enzimas de Conjugação de Ubiquitina/metabolismo
8.
Mol Immunol ; 45(7): 1847-62, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18083234

RESUMO

AIRE (autoimmune regulator) promotes the establishment of self-tolerance by regulating gene expression in the thymus. Mutations in AIRE lead to an autoimmune disease, APECED. Here we have identified PIAS proteins as novel AIRE interaction partners. Although PIAS proteins function as E3 SUMO ligases, AIRE is not sumoylated. We expressed AIRE, wt PIAS1, and PIAS1 mutants with deleted SP-RING domain or SUMO interaction motif (SIM) in different cell lines and demonstrate that AIRE and PIAS1 localize to adjacent nuclear bodies (NBs). The expression of AIRE enhances the formation of PIAS1 NBs. The ability of PIAS1 to localize into NBs and interconnect with AIRE is neither dependent on the SP-RING domain nor the SIM. Further, we show that PIAS1 is able to attract AIRE into SUMO1-containing complexes and that the process is dependent on the SIM of PIAS1. PIAS1 and AIRE concurrently activate the human insulin promoter, a known target gene of AIRE, and the SP-RING is required for this activation. Moreover, AIRE represses and PIAS1 activates the CSTB promoter, used as a model for a housekeeping promoter, and both the SP-RING and SIM are needed for its activation by PIAS1. Collectively, our data suggest that AIRE and PIAS1 interact functionally to regulate the activities of the target genes of AIRE.


Assuntos
Regulação da Expressão Gênica , Proteínas Inibidoras de STAT Ativados/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Motivos de Aminoácidos , Animais , Linhagem Celular , Cistatina B , Cistatinas/genética , Humanos , Insulina/genética , Camundongos , Proteínas Mutantes/metabolismo , Matriz Nuclear/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/química , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Transporte Proteico , Deleção de Sequência , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Frações Subcelulares/metabolismo , Ativação Transcricional/genética , Transfecção , Proteína AIRE
9.
Dis Model Mech ; 12(1)2019 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-30559154

RESUMO

Perturbation of protein homeostasis and aggregation of misfolded proteins is a major cause of many human diseases. A hallmark of the neurodegenerative disease spinocerebellar ataxia type 7 (SCA7) is the intranuclear accumulation of mutant, misfolded ataxin-7 (polyQ-ATXN7). Here, we show that endogenous ATXN7 is modified by SUMO proteins, thus also suggesting a physiological role for this modification under conditions of proteotoxic stress caused by the accumulation of polyQ-ATXN7. Co-immunoprecipitation experiments, immunofluorescence microscopy and proximity ligation assays confirmed the colocalization and interaction of polyQ-ATXN7 with SUMO2 in cells. Moreover, upon inhibition of the proteasome, both endogenous SUMO2/3 and the RNF4 ubiquitin ligase surround large polyQ-ATXN7 intranuclear inclusions. Overexpression of RNF4 and/or SUMO2 significantly decreased levels of polyQ-ATXN7 and, upon proteasomal inhibition, led to a marked increase in the polyubiquitination of polyQ-ATXN7. This provides a mechanism for the clearance of polyQ-ATXN7 from affected cells that involves the recruitment of RNF4 by SUMO2/3-modified polyQ-ATXN7, thus leading to its ubiquitination and proteasomal degradation. In a SCA7 knock-in mouse model, we similarly observed colocalization of SUMO2/3 with polyQ-ATXN7 inclusions in the cerebellum and retina. Furthermore, we detected accumulation of SUMO2/3 high-molecular-mass species in the cerebellum of SCA7 knock-in mice, compared with their wild-type littermates, and changes in SUMO-related transcripts. Immunohistochemical analysis showed the accumulation of SUMO proteins and RNF4 in the cerebellum of SCA7 patients. Taken together, our results show that the SUMO pathway contributes to the clearance of aggregated ATXN7 and suggest that its deregulation might be associated with SCA7 disease progression.


Assuntos
Ataxina-7/metabolismo , Proteínas Nucleares/metabolismo , Dobramento de Proteína , Proteólise , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ataxias Espinocerebelares/metabolismo , Sumoilação , Fatores de Transcrição/metabolismo , Animais , Cerebelo/metabolismo , Criança , Modelos Animais de Doenças , Células HEK293 , Células HeLa , Humanos , Corpos de Inclusão/metabolismo , Células MCF-7 , Camundongos , Pessoa de Meia-Idade , Mutação/genética , Proteína da Leucemia Promielocítica/metabolismo , Inibidores de Proteassoma/farmacologia , Agregados Proteicos/efeitos dos fármacos , Dobramento de Proteína/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Ataxias Espinocerebelares/patologia , Sumoilação/efeitos dos fármacos , Ubiquitina/metabolismo
10.
Cell Stem Cell ; 23(5): 742-757.e8, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30401455

RESUMO

Understanding general principles that safeguard cellular identity should reveal critical insights into common mechanisms underlying specification of varied cell types. Here, we show that SUMO modification acts to stabilize cell fate in a variety of contexts. Hyposumoylation enhances pluripotency reprogramming in vitro and in vivo, increases lineage transdifferentiation, and facilitates leukemic cell differentiation. Suppressing sumoylation in embryonic stem cells (ESCs) promotes their conversion into 2-cell-embryo-like (2C-like) cells. During reprogramming to pluripotency, SUMO functions on fibroblastic enhancers to retain somatic transcription factors together with Oct4, Sox2, and Klf4, thus impeding somatic enhancer inactivation. In contrast, in ESCs, SUMO functions on heterochromatin to silence the 2C program, maintaining both proper H3K9me3 levels genome-wide and repression of the Dux locus by triggering recruitment of the sumoylated PRC1.6 and Kap/Setdb1 repressive complexes. Together, these studies show that SUMO acts on chromatin as a glue to stabilize key determinants of somatic and pluripotent states.


Assuntos
Cromatina/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Animais , Células Cultivadas , Reprogramação Celular , Fator 4 Semelhante a Kruppel , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição/metabolismo
11.
Nat Rev Cancer ; 17(3): 184-197, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28134258

RESUMO

Post-translational protein modification by small ubiquitin-like modifier (SUMO), termed sumoylation, is an important mechanism in cellular responses to stress and one that appears to be upregulated in many cancers. Here, we examine the role of sumoylation in tumorigenesis as a possibly necessary safeguard that protects the stability and functionality of otherwise easily misregulated gene expression programmes and signalling pathways of cancer cells.


Assuntos
Neoplasias/etiologia , Proteína SUMO-1/fisiologia , Animais , Hipóxia Celular , Senescência Celular , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Regulação da Expressão Gênica , Humanos , NF-kappa B/fisiologia , Transdução de Sinais/fisiologia , Sumoilação
12.
Mol Cell Biol ; 33(11): 2163-77, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23530056

RESUMO

Arkadia is a RING domain E3 ubiquitin ligase that activates the transforming growth factor ß (TGF-ß) pathway by inducing degradation of the inhibitor SnoN/Ski. Here we show that Arkadia contains three successive SUMO-interacting motifs (SIMs) that mediate noncovalent interaction with poly-SUMO2. We identify the third SIM (VVDL) of Arkadia to be the most relevant one in this interaction. Furthermore, we provide evidence that Arkadia can function as a SUMO-targeted ubiquitin ligase (STUBL) by ubiquitinating SUMO chains. While the SIMs of Arkadia are not essential for SnoN/Ski degradation in response to TGF-ß, we show that they are necessary for the interaction of Arkadia with polysumoylated PML in response to arsenic and its concomitant accumulation into PML nuclear bodies. Moreover, Arkadia depletion leads to accumulation of polysumoylated PML in response to arsenic, highlighting a requirement of Arkadia for arsenic-induced degradation of polysumoylated PML. Interestingly, Arkadia homodimerizes but does not heterodimerize with RNF4, the other STUBL involved in PML degradation, suggesting that these two E3 ligases do not act synergistically but most probably act independently during this process. Altogether, these results identify Arkadia to be a novel STUBL that can trigger degradation of signal-induced polysumoylated proteins.


Assuntos
Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Arsênio/farmacologia , Linhagem Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases/genética
14.
Proc Natl Acad Sci U S A ; 104(3): 893-8, 2007 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-17209013

RESUMO

The sheltering of chromosome ends from illegitimate DNA repair reactions and telomere length homeostasis are critical for preserving genomic integrity. Growing evidence implicates covalent protein modification by SUMO (small ubiquitin-like modifier) (sumoylation) in the regulation of numerous DNA transactions, including DNA repair and transcription, as well as heterochromatin formation and maintenance. We have recently shown that fission yeast Pli1p is a SUMO E3 ligase and that pli1 mutants, which are impaired for global sumoylation, are viable, but exhibit de-regulated homologous recombination and marked defects in chromosome segregation and centromeric silencing, as well as a consistent increase in telomere length. In this work, we explore the mechanisms underlying sumoylation-dependent telomere maintenance. We show that Pli1p, but not the related Nse2p, is the principal SUMO E3 ligase enzyme involved. Using both a pli1 mutation and a physiological "knockdown" of sumoylation, achieved by inducible expression of a dominant negative form of the conjugating enzyme Ubc9p, we further show that telomere lengthening induced by lack of sumoylation is not due to unscheduled telomere-telomere recombination. Instead, sumoylation increases telomerase activity, therefore suggesting that this modification controls the activity of a positive or negative regulator of telomerase.


Assuntos
Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Telômero/genética , Telômero/metabolismo , DNA Recombinante/genética , Mutação/genética , Ligação Proteica , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
15.
Nat Rev Mol Cell Biol ; 4(9): 690-9, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14506472

RESUMO

Post-translational modification by the ubiquitin-like SUMO protein is emerging as a defining feature of eukaryotic cells. Sumoylation has crucial roles in the regulatory challenges that face nucleate cells, including the control of nucleocytoplasmic signalling and transport and the faithful replication of a large and complex genome, as well as the regulation of gene expression.


Assuntos
Núcleo Celular/metabolismo , Proteína SUMO-1/fisiologia , Animais , Humanos , Transcrição Gênica/fisiologia
16.
EMBO J ; 23(19): 3844-53, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15359282

RESUMO

Sumoylation represents a conserved mechanism of post-translational protein modification. We report that Pli1p, the unique fission yeast member of the SP-RING family, is a SUMO E3 ligase in vivo and in vitro. pli1Delta cells display no obvious mitotic growth defects, but are sensitive to the microtubule-destabilizing drug TBZ and exhibit enhanced minichromosome loss. The weakened centromeric function of pli1Delta cells may be related to the defective heterochromatin structure at the central core, as shown by the reduced silencing of an ura4 variegation reporter gene inserted at cnt and imr. Interestingly, pli1Delta cells also exhibit enhanced loss of the ura4 reporter at these loci, likely by gene conversion using homologous sequences as information donors. Moreover, pli1Delta cells exhibit consistent telomere length increase, possibly achieved by a similar process. Point mutations within the RING finger of Pli1p totally or partially reproduce the pli1 deletion phenotypes, thus correlating with their sumoylation activity. Altogether, these results strongly suggest that Pli1p, and by extension sumoylation, is involved in mechanisms that regulate recombination in particular heterochromatic repeated sequences.


Assuntos
Centrômero/fisiologia , Heterocromatina/fisiologia , Proteínas de Schizosaccharomyces pombe/fisiologia , Schizosaccharomyces/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Telômero/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Sequência de Aminoácidos , Cromossomos Fúngicos/metabolismo , Genes Reporter/fisiologia , Ligases , Mitose/efeitos dos fármacos , Dados de Sequência Molecular , Fenótipo , Mutação Puntual/genética , Recombinação Genética , Deleção de Sequência , Homologia de Sequência de Aminoácidos
17.
EMBO Rep ; 3(10): 975-81, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12231507

RESUMO

In mammalian cells, as in Schizosaccharomyces pombe and Drosophila, HP1 proteins bind histone H3 tails methylated on lysine 9 (K9). However, whereas K9-methylated H3 histones are distributed throughout the nucleus, HP1 proteins are enriched in pericentromeric heterochromatin. This observation suggests that the methyl-binding property of HP1 may not be sufficient for its heterochromatin targeting. We show that the association of HP1alpha with pericentromeric heterochromatin depends not only on its methyl-binding chromo domain but also on an RNA-binding activity present in the hinge region of the protein that connects the conserved chromo and chromoshadow domains. Our data suggest the existence of complex heterochromatin binding sites composed of methylated histone H3 tails and RNA, with each being recognized by a separate domain of HP1alpha.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Heterocromatina/metabolismo , RNA/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Western Blotting , Núcleo Celular/metabolismo , Cromatina/metabolismo , Homólogo 5 da Proteína Cromobox , Drosophila , Eletroforese em Gel de Poliacrilamida , Escherichia coli/metabolismo , Vetores Genéticos , Glutationa Transferase/metabolismo , Imuno-Histoquímica , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos
18.
Cell ; 108(1): 109-20, 2002 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-11792325

RESUMO

Posttranslational modification with SUMO1 regulates protein/protein interactions, localization, and stability. SUMOylation requires the E1 enzyme Aos1/Uba2 and the E2 enzyme Ubc9. A family of E3-like factors, PIAS proteins, was discovered recently. Here we show that the nucleoporin RanBP2/Nup358 also has SUMO1 E3-like activity. RanBP2 directly interacts with the E2 enzyme Ubc9 and strongly enhances SUMO1-transfer from Ubc9 to the SUMO1 target Sp100. The E3-like activity is contained within a 33 kDa domain of RanBP2 that lacks RING finger motifs and does not resemble PIAS family proteins. Our findings place SUMOylation at the cytoplasmic filaments of the NPC and suggest that, at least for some substrates, modification and nuclear import are linked events.


Assuntos
Antígenos Nucleares , Ligases/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteína SUMO-1/metabolismo , Enzimas de Conjugação de Ubiquitina , Animais , Anticorpos , Autoantígenos/metabolismo , Proteínas de Bactérias/metabolismo , Núcleo Celular/metabolismo , Citoplasma/enzimologia , Proteínas Ativadoras de GTPase/imunologia , Proteínas Ativadoras de GTPase/metabolismo , Cabras , Proteínas Luminescentes/metabolismo , Chaperonas Moleculares , Complexo de Proteínas Formadoras de Poros Nucleares/imunologia , Proteínas Nucleares/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Coelhos , Ubiquitina-Proteína Ligases , Dedos de Zinco/fisiologia
19.
EMBO J ; 21(11): 2682-91, 2002 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-12032081

RESUMO

Transcriptional repression mediated through histone deacetylation is a critical component of eukaryotic gene regulation. Here we demonstrate that the class II histone deacetylase HDAC4 is covalently modified by the ubiquitin-related SUMO-1 modifier. A sumoylation-deficient point mutant (HDAC4-K559R) shows a slightly impaired ability to repress transcription as well as reduced histone deacetylase activity. The ability of HDAC4 to self-aggregate is a prerequisite for proper sumoylation in vivo. Calcium/calmodulin-dependent protein kinase (CaMK) signalling, which induces nuclear export, abrogates SUMO-1 modification of HDAC4. Moreover, the modification depends on the presence of an intact nuclear localization signal and is catalysed by the nuclear pore complex (NPC) RanBP2 protein, a factor newly identified as a SUMO E3 ligase. These findings suggest that sumoylation of HDAC4 takes place at the NPC and is coupled to its nuclear import. Finally, modification experiments indicate that the MEF2-interacting transcription repressor (MITR) as well as HDAC1 and -6 are similarly SUMO modified, indicating that sumoylation may be an important regulatory mechanism for the control of transcriptional repression mediated by both class I and II HDACs.


Assuntos
Histona Desacetilases/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Proteínas Repressoras/metabolismo , Animais , Western Blotting , Células COS , Núcleo Celular/metabolismo , Cromatina/metabolismo , Citoplasma/metabolismo , Dimerização , Eletroforese em Gel de Poliacrilamida , Genes Reporter , Glutationa Transferase/metabolismo , Células HeLa , Humanos , Lisina/química , Microscopia de Fluorescência , Chaperonas Moleculares , Mutação , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA