Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
2.
Biochem Biophys Res Commun ; 633: 61-63, 2022 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-36344165

RESUMO

Ubiquitin-fold modifier 1 (UFM1) is a newly identified ubiquitin-like protein. Like ubiquitin, UFM1 is conjugated to its target proteins through a three-step enzyme system: UBA5 (E1), UFC1 (E2), and UFL1 (E3), but with an additional essential component, UFBP1. This protein modification by UFM1 (ufmylation) can be reversed by UFM1-specific proteases (UFSPs). So far only a handful of target proteins for ufmylation have been identified, and they are mostly associated with either promotion or suppression of tumorigenesis. Here, we summarize the recent progress in the knowledge of tumor-suppressive and tumorigenic functions of ufmylation as well as in the development of therapeutic drugs against ufmylation-associated cancer.


Assuntos
Neoplasias , Processamento de Proteína Pós-Traducional , Humanos , Enzimas Ativadoras de Ubiquitina/genética , Proteínas/metabolismo , Neoplasias/metabolismo , Ubiquitinas/metabolismo
3.
Mol Cell ; 54(4): 626-38, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-24768535

RESUMO

In response to DNA damage, PCNA is mono-ubiquitinated and triggers translesion DNA synthesis (TLS) by recruiting polymerase-η. However, it remained unknown how error-prone TLS is turned off after DNA lesion bypass to prevent mutagenesis. Here we showed that ISG15 modification (ISGylation) of PCNA plays a key role in TLS termination. Upon UV irradiation, EFP, an ISG15 E3 ligase, bound to mono-ubiquitinated PCNA and promoted its ISGylation. ISGylated PCNA then tethered USP10 for deubiquitination and in turn the release of polymerase-η from PCNA. Eventually, PCNA was deISGylated by UBP43 for reloading of replicative DNA polymerases and resuming normal DNA replication. However, ISGylation-defective Lys-to-Arg mutations in PCNA or knockdown of any of ISG15, EFP, or USP10 led to persistent recruitment of mono-ubiquitinated PCNA and polymerase-η to nuclear foci, causing an increase in mutation frequency. These findings establish a crucial role of PCNA ISGylation in termination of error-prone TLS for preventing excessive mutagenesis.


Assuntos
Citocinas/metabolismo , Dano ao DNA , Replicação do DNA , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ubiquitinas/metabolismo , Arginina/metabolismo , Sítios de Ligação/genética , Citocinas/genética , DNA Polimerase II/metabolismo , Reparo do DNA , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Lisina/metabolismo , Mutagênese , Taxa de Mutação , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Ubiquitinas/genética
4.
Mol Cell ; 56(2): 261-274, 2014 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-25219498

RESUMO

Biological roles for UFM1, a ubiquitin-like protein, are largely unknown, and therefore we screened for targets of ufmylation. Here we show that ufmylation of the nuclear receptor coactivator ASC1 is a key step for ERα transactivation in response to 17ß-estradiol (E2). In the absence of E2, the UFM1-specific protease UfSP2 was bound to ASC1, which maintains ASC1 in a nonufmylated state. In the presence of E2, ERα bound ASC1 and displaced UfSP2, leading to ASC1 ufmylation. Polyufmylation of ASC1 enhanced association of p300, SRC1, and ASC1 at promoters of ERα target genes. ASC1 overexpression or UfSP2 knockdown promoted ERα-mediated tumor formation in vivo, which could be abrogated by treatment with the anti-breast cancer drug tamoxifen. In contrast, expression of ufmylation-deficient ASC1 mutant or knockdown of the UFM1-activating E1 enzyme UBA5 prevented tumor growth. These findings establish a role for ASC1 ufmylation in breast cancer development by promoting ERα transactivation.


Assuntos
Sistema y+ de Transporte de Aminoácidos/metabolismo , Neoplasias da Mama/patologia , Receptor alfa de Estrogênio/metabolismo , Proteínas/química , Sistema y+ de Transporte de Aminoácidos/química , Sistema y+ de Transporte de Aminoácidos/genética , Animais , Neoplasias da Mama/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Cisteína Endopeptidases/metabolismo , Proteína p300 Associada a E1A/genética , Ativação Enzimática/genética , Estradiol/genética , Estradiol/metabolismo , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/genética , Feminino , Células HEK293 , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Coativador 1 de Receptor Nuclear/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Proteínas/metabolismo , Tamoxifeno/farmacologia , Ativação Transcricional , Ubiquitina/metabolismo , Enzimas Ativadoras de Ubiquitina/genética , Ubiquitina-Proteína Ligases/metabolismo
5.
Mol Cell ; 48(4): 572-86, 2012 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-23063525

RESUMO

Ubiquitination plays a major role in protein degradation. Although phosphorylation-dependent ubiquitination is well known for the regulation of protein stability, methylation-dependent ubiquitination machinery has not been characterized. Here, we provide evidence that methylation-dependent ubiquitination is carried out by damage-specific DNA binding protein 1 (DDB1)/cullin4 (CUL4) E3 ubiquitin ligase complex and a DDB1-CUL4-associated factor 1 (DCAF1) adaptor, which recognizes monomethylated substrates. Molecular modeling and binding affinity studies reveal that the putative chromo domain of DCAF1 directly recognizes monomethylated substrates, whereas critical binding pocket mutations of the DCAF1 chromo domain ablated the binding from the monomethylated substrates. Further, we discovered that enhancer of zeste homolog 2 (EZH2) methyltransferase has distinct substrate specificities for histone H3K27 and nonhistones exemplified by an orphan nuclear receptor, RORα. We propose that EZH2-DCAF1/DDB1/CUL4 represents a previously unrecognized methylation-dependent ubiquitination machinery specifically recognizing "methyl degron"; through this, nonhistone protein stability can be dynamically regulated in a methylation-dependent manner.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Culina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste , Humanos , Células MCF-7 , Metilação , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Proteínas Serina-Treonina Quinases , Especificidade por Substrato
6.
EMBO J ; 31(23): 4441-52, 2012 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-23092970

RESUMO

Heterogeneous ribonucleoprotein-K (hnRNP-K) is normally ubiquitinated by HDM2 for proteasome-mediated degradation. Under DNA-damage conditions, hnRNP-K is transiently stabilized and serves as a transcriptional co-activator of p53 for cell-cycle arrest. However, how the stability and function of hnRNP-K is regulated remained unknown. Here, we demonstrated that UV-induced SUMOylation of hnRNP-K prevents its ubiquitination for stabilization. Using SUMOylation-defective mutant and purified SUMOylated hnRNP-K, SUMOylation was shown to reduce hnRNP-K's affinity to HDM2 with an increase in that to p53 for p21-mediated cell-cycle arrest. PIAS3 served as a small ubiquitin-related modifier (SUMO) E3 ligase for hnRNP-K in an ATR-dependent manner. During later periods after UV exposure, however, SENP2 removed SUMO from hnRNP-K for its destabilization and in turn for release from cell-cycle arrest. Consistent with the rise-and-fall of both SUMOylation and stability of hnRNP-K, its ability to interact with PIAS3 was inversely correlated to that with SENP2 during the time course after UV exposure. These findings indicate that SUMO modification plays a crucial role in the control of hnRNP-K's function as a p53 co-activator in response to DNA damage by UV.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Cisteína Endopeptidases/metabolismo , Dano ao DNA , Células HEK293 , Células HeLa , Humanos , Modelos Biológicos , Chaperonas Moleculares/metabolismo , Mutação , Proteínas Inibidoras de STAT Ativados/metabolismo , Sumoilação , Ubiquitina/química , Raios Ultravioleta
7.
J Neurosci ; 33(31): 12728-38, 2013 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-23904609

RESUMO

Katanin is a heterodimeric enzyme that severs and disassembles microtubules. While the p60 subunit has the enzyme activity, the p80 subunit regulates the p60 activity. The microtubule-severing activity of katanin plays an essential role in axonal growth. However, the mechanisms by which neuronal cells regulate the expression of katanin-p60 remains unknown. Here we showed that USP47 and C terminus of Hsp70-interacting protein (CHIP) antagonistically regulate the stability of katanin-p60 and thereby axonal growth. USP47 was identified as a katanin-p60-specific deubiquitinating enzyme for its stabilization. We also identified CHIP as a ubiquitin E3 ligase that promotes proteasome-mediated degradation of katanin-p60. Moreover, USP47 promoted axonal growth of cultured rat hippocampal neurons, whereas CHIP inhibited it. Significantly, treatment with basic fibroblast growth factor (bFGF), an inducer of axonal growth, increased the levels of USP47 and katanin-p60, but not CHIP. Consistently, bFGF treatment resulted in a marked decrease in the level of ubiquitinated katanin-p60 and thereby in the promotion of axonal growth. On the other hand, the level of USP47, but not CHIP, decreased concurrently with that of katanin-p60 as axons reached their target cells. These results indicate that USP47 plays a crucial role in the control of axonal growth during neuronal development by antagonizing CHIP-mediated katanin-p60 degradation.


Assuntos
Adenosina Trifosfatases/metabolismo , Axônios/fisiologia , Proteínas de Choque Térmico HSP47/metabolismo , Neurônios/citologia , Ubiquitina-Proteína Ligases/metabolismo , Adenosina Trifosfatases/genética , Animais , Células Cultivadas , Embrião de Mamíferos , Feminino , Fatores de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Choque Térmico HSP47/genética , Hipocampo/citologia , Humanos , Imunoprecipitação , Katanina , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Transfecção , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/genética
9.
Biochem Biophys Res Commun ; 455(3-4): 153-8, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25450678

RESUMO

c-Cbl, a RING-type ubiquitin E3 ligase, down-regulates receptor tyrosine kinases, including EGF receptor, and inhibits cell proliferation. Moreover, c-Cbl mutations are frequently found in patients with myeloid neoplasm. Therefore, c-Cbl is known as a tumor suppressor. αPix is expressed only in highly proliferative and mobile cells, including immune cells, and up-regulated in certain invasive tumors, such as glioblastoma multiforme. Here, we showed that c-Cbl serves as an ubiquitin E3 ligase for proteasome-mediated degradation of αPix, but not ßPix. Remarkably, the rat C6 and human A172 glioma cells were unable to express c-Cbl, which leads to a dramatic accumulation of αPix. Depletion of αPix by shRNA markedly reduced the ability of the glioma cells to migrate and invade, whereas complementation of shRNA-insensitive αPix promoted it. These results indicate that c-Cbl negatively regulates αPix-mediated cell migration and invasion and the lack of c-Cbl in the C6 and A172 glioma cells is responsible for their malignant behavior.


Assuntos
Leucemia Mieloide/genética , Mutação , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Receptores ErbB/metabolismo , Teste de Complementação Genética , Glioma/metabolismo , Células HEK293 , Humanos , Leucemia Mieloide/metabolismo , Invasividade Neoplásica , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Cima
10.
Nat Cell Biol ; 8(6): 631-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16699503

RESUMO

Defining the functional modules within transcriptional regulatory factors that govern switching between repression and activation events is a central issue in biology. Recently, we have reported the dynamic role of a beta-catenin-reptin chromatin remodelling complex in regulating a metastasis suppressor gene KAI1 (ref.1), which is capable of inhibiting the progression of tumour metastasis. Here, we identify signalling factors that confer repressive function on reptin and hence repress the expression of KAI1. Biochemical purification of a reptin-containing complex has revealed the presence of specific desumoylating enzymes that reverse the sumoylation of reptin that underlies its function as a repressor. Desumoylation of reptin alters the repressive function of reptin and its association with HDAC1. Furthermore, the sumoylation status of reptin modulates the invasive activity of cancer cells with metastatic potential. These data clearly define a functional model and provide a novel link for SUMO modification in cancer metastasis.


Assuntos
Proteínas de Transporte/fisiologia , Cromatina/metabolismo , DNA Helicases/fisiologia , Metástase Neoplásica , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , DNA Helicases/metabolismo , Regulação da Expressão Gênica , Histona Desacetilase 1 , Histona Desacetilases/metabolismo , Humanos , Proteína Kangai-1/genética , Ligação Proteica , Proteínas Repressoras , Transdução de Sinais
11.
Nat Cell Biol ; 8(12): 1424-31, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17086174

RESUMO

The p53 tumour suppressor has a key role in the control of cell growth and differentiation, and in the maintenance of genome integrity. p53 is kept labile under normal conditions, but in response to stresses, such as DNA damage, it accumulates in the nucleus for induction of cell-cycle arrest, DNA repair or apoptosis. Mdm2 is an ubiquitin ligase that promotes p53 ubiquitination and degradation. Mdm2 is also self-ubiquitinated and degraded. Here, we identified a novel cascade for the increase in p53 level in response to DNA damage. A new SUMO-specific protease, SUSP4, removed SUMO-1 from Mdm2 and this desumoylation led to promotion of Mdm2 self-ubiquitination, resulting in p53 stabilization. Moreover, SUSP4 competed with p53 for binding to Mdm2, also resulting in p53 stabilization. Overexpression of SUSP4 inhibited cell growth, whereas knockdown of susp4 by RNA interference (RNAi) promoted of cell growth. UV damage induced SUSP4 expression, leading to an increase in p53 levels in parallel with a decrease in Mdm2 levels. These findings establish a new mechanism for the elevation of cellular p53 levels in response to UV damage.


Assuntos
Cisteína Endopeptidases/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina/metabolismo , Animais , Processos de Crescimento Celular/efeitos da radiação , Cisteína Endopeptidases/genética , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Células NIH 3T3 , Ligação Proteica/efeitos da radiação , Transporte Proteico/efeitos da radiação , Proteínas Proto-Oncogênicas c-mdm2/deficiência , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Termodinâmica , Proteína Supressora de Tumor p53/deficiência , Raios Ultravioleta
12.
J Biol Chem ; 286(12): 10248-57, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21228277

RESUMO

Ubiquitin-fold modifier 1 (Ufm1)-specific protease 2 (UfSP2) is a cysteine protease that is responsible for the release of Ufm1 from Ufm1-conjugated cellular proteins, as well as for the generation of mature Ufm1 from its precursor. The 2.6 Å resolution crystal structure of mouse UfSP2 reveals that it is composed of two domains. The C-terminal catalytic domain is similar to UfSP1 with Cys(294), Asp(418), His(420), Tyr(282), and a regulatory loop participating in catalysis. The novel N-terminal domain shows a unique structure and plays a role in the recognition of its cellular substrate C20orf116 and thus in the recruitment of UfSP2 to the endoplasmic reticulum, where C20orf116 predominantly localizes. Mutagenesis studies were carried out to provide the structural basis for understanding the loss of catalytic activity observed in a recently identified UfSP2 mutation that is associated with an autosomal dominant form of hip dysplasia.


Assuntos
Cisteína Endopeptidases/química , Proteínas , Animais , Catálise , Cristalografia por Raios X , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/genética , Luxação Congênita de Quadril/enzimologia , Luxação Congênita de Quadril/genética , Humanos , Camundongos , Estrutura Terciária de Proteína
13.
J Biol Chem ; 286(28): 24743-53, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21586565

RESUMO

Bax inhibitor-1 (BI-1) is an evolutionarily conserved protein that protects cells against endoplasmic reticulum (ER) stress while also affecting the ER stress response. In this study, we examined BI-1-induced regulation of the ER stress response as well as the control of the protein over cell death under ER stress. In BI-1-overexpressing cells (BI-1 cells), proteasome activity was similar to that of control cells; however, the lysosomal fraction of BI-1 cells showed sensitivity to degradation of BSA. In addition, areas and polygonal lengths of lysosomes were greater in BI-1 cells than in control cells, as assessed by fluorescence and electron microscopy. In BI-1 cells, lysosomal pH was lower than in control cells and lysosomal vacuolar H(+)-ATPase(V-ATPase), a proton pump, was activated, suggesting high H(+) uptake into lysosomes. Even when exposed to ER stress, BI-1 cells maintained high levels of lysosomal activities, including V-ATPase activity. Bafilomycin, a V-ATPase inhibitor, leads to the reversal of BI-1-induced regulation of ER stress response and cell death due to ER stress. In BI-1 knock-out mouse embryo fibroblasts, lysosomal activity and number per cell were relatively lower than in BI-1 wild-type cells. This study suggests that highly maintained lysosomal activity may be one of the mechanisms by which BI-1 exerts its regulatory effects on the ER stress response and cell death.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Lisossomos/metabolismo , Proteínas de Membrana/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Morte Celular/fisiologia , Linhagem Celular Tumoral , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Lisossomos/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , ATPases Vacuolares Próton-Translocadoras/genética , ATPases Vacuolares Próton-Translocadoras/metabolismo
14.
Mol Cells ; 45(6): 425-434, 2022 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-35680375

RESUMO

The post-translational modification (e.g., phosphorylation) of estrogen receptor α (ERα) plays a role in controlling the expression and subcellular localization of ERα as well as its sensitivity to hormone response. Here, we show that ERα is also modified by UFM1 and this modification (ufmylation) plays a crucial role in promoting the stability and transactivity of ERα, which in turn promotes breast cancer development. The elevation of ufmylation via the knockdown of UFSP2 (the UFM1-deconjugating enzyme in humans) dramatically increases ERα stability by inhibiting ubiquitination. In contrast, ERα stability is decreased by the prevention of ufmylation via the silencing of UBA5 (the UFM1-activating E1 enzyme). Lys171 and Lys180 of ERα were identified as the major UFM1 acceptor sites, and the replacement of both Lys residues by Arg (2KR mutation) markedly reduced ERα stability. Moreover, the 2KR mutation abrogated the 17ß-estradiol-induced transactivity of ERα and the expression of its downstream target genes, including pS2, cyclin D1, and c-Myc; this indicates that ERα ufmylation is required for its transactivation function. In addition, the 2KR mutation prevented anchorage-independent colony formation by MCF7 cells. Most notably, the expression of UFM1 and its conjugating machinery (i.e., UBA5, UFC1, UFL1, and UFBP1) were dramatically upregulated in ERα-positive breast cancer cell lines and tissues. Collectively, these findings implicate a critical role attributed to ERα ufmylation in breast cancer development by ameliorating its stability and transactivity.


Assuntos
Neoplasias da Mama , Receptor alfa de Estrogênio , Neoplasias da Mama/genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Células MCF-7 , Proteínas/química , Enzimas Ativadoras de Ubiquitina/química , Enzimas Ativadoras de Ubiquitina/genética , Enzimas Ativadoras de Ubiquitina/metabolismo , Ubiquitinação
15.
J Biol Chem ; 285(8): 5417-27, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20018847

RESUMO

The ubiquitin fold modifier 1 (Ufm1) is the most recently discovered ubiquitin-like modifier whose conjugation (ufmylation) system is conserved in multicellular organisms. Ufm1 is known to covalently attach with cellular protein(s) via a specific E1-activating enzyme (Uba5) and an E2-conjugating enzyme (Ufc1), but its E3-ligating enzyme(s) as well as the target protein(s) remain unknown. Herein, we report both a novel E3 ligase for Ufm1, designated Ufl1, and an Ufm1-specific substrate ligated by Ufl1, C20orf116. Ufm1 was covalently conjugated with C20orf116. Although Ufl1 has no obvious sequence homology to any other known E3s for ubiquitin and ubiquitin-like modifiers, the C20orf116 x Ufm1 formation was greatly accelerated by Ufl1. The C20orf116 x Ufm1 conjugate was cleaved by Ufm1-specific proteases, implying the reversibility of ufmylation. The conjugation was abundant in the liver and lungs of Ufm1-transgenic mice, fractionated into membrane fraction, and impaired in Uba5 knock-out cells. Intriguingly, immunological analysis revealed localizations of Ufl1 and C20orf116 mainly to the endoplasmic reticulum. Our results provide novel insights into the Ufm1 system involved in cellular regulation of multicellular organisms.


Assuntos
Proteínas de Transporte/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Animais , Proteínas de Transporte/genética , Retículo Endoplasmático/genética , Humanos , Camundongos , Camundongos Knockout , Proteínas/genética , Ubiquitina/genética , Enzimas Ativadoras de Ubiquitina/genética , Enzimas Ativadoras de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética
16.
Biochim Biophys Acta ; 1802(5): 485-96, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20153823

RESUMO

ISG15, the product of interferon (IFN)-stimulated gene 15, is the first identified ubiquitin-like protein, consisting of two ubiquitin-like domains. ISG15 is synthesized as a precursor in certain mammals and, therefore, needs to be processed to expose the C-terminal glycine residue before conjugation to target proteins. A set of three-step cascade enzymes, an E1 enzyme (UBE1L), an E2 enzyme (UbcH8), and one of several E3 ligases (e.g., EFP and HERC5), catalyzes ISG15 conjugation (ISGylation) of a specific protein. These enzymes are unique among the cascade enzymes for ubiquitin and other ubiquitin-like proteins in that all of them are induced by type I IFNs or other stimuli, such as exposure to viruses and lipopolysaccharide. Mass spectrometric analysis has led to the identification of several hundreds of candidate proteins that can be conjugated by ISG15. Some of them are type I IFN-induced proteins, such as PKR and RIG-I, and some are the key regulators that are involved in IFN signaling, such as JAK1 and STAT1, implicating the role of ISG15 and its conjugates in type I IFN-mediated innate immune responses. However, relatively little is known about the functional significance of ISG15 induction due to the lack of information on the consequences of its conjugation to target proteins. Here, we describe the recent progress made in exploring the biological function of ISG15 and its reversible modification of target proteins and thus in their implication in immune diseases.


Assuntos
Citocinas/fisiologia , Doenças do Sistema Imunitário/imunologia , Ubiquitinas/fisiologia , Humanos
17.
EMBO Rep ; 10(4): 374-80, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19270716

RESUMO

Interferon (IFN)-induced signalling pathways have essential functions in innate immune responses. In response to type I IFNs, filamin B tethers RAC1 and a Jun N-terminal kinase (JNK)-specific mitogen-activated protein kinase (MAPK) module--MEKK1, MKK4 and JNK--and thereby promotes the activation of JNK and JNK-mediated apoptosis. Here, we show that type I IFNs induce the conjugation of filamin B by interferon-stimulated gene 15 (ISG15). ISGylation of filamin B led to the release of RAC1, MEKK1 and MKK4 from the scaffold protein and thus to the prevention of sequential activation of the JNK cascade. By contrast, blockade of filamin B ISGylation by substitution of Lys 2467 with arginine or by knockdown of ubiquitin-activating enzyme E1-like (UBEL1) prevented the release of the signalling molecules from filamin B, resulting in persistent promotion of JNK activation and JNK-mediated apoptosis. These results indicate that filamin B ISGylation acts as a negative feedback regulatory gate for the desensitization of type I IFN-induced JNK signalling.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas Contráteis/metabolismo , Citocinas/metabolismo , Interferon Tipo I/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas dos Microfilamentos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ubiquitinas/metabolismo , Linhagem Celular , Filaminas , Células HeLa , Humanos , Imunoprecipitação , Modelos Biológicos , Enzimas de Conjugação de Ubiquitina/metabolismo
18.
Nature ; 434(7035): 921-6, 2005 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-15829968

RESUMO

Defining the molecular strategies that integrate diverse signalling pathways in the expression of specific gene programmes that are critical in homeostasis and disease remains a central issue in biology. This is particularly pertinent in cancer biology because downregulation of tumour metastasis suppressor genes is a common occurrence, and the underlying molecular mechanisms are not well established. Here we report that the downregulation of a metastasis suppressor gene, KAI1, in prostate cancer cells involves the inhibitory actions of beta-catenin, along with a reptin chromatin remodelling complex. This inhibitory function of beta-catenin-reptin requires both increased beta-catenin expression and recruitment of histone deacetylase activity. The coordinated actions of beta-catenin-reptin components that mediate the repressive state serve to antagonize a Tip60 coactivator complex that is required for activation; the balance of these opposing complexes controls the expression of KAI1 and metastatic potential. The molecular mechanisms underlying the antagonistic regulation of beta-catenin-reptin and the Tip60 coactivator complexes for the metastasis suppressor gene, KAI1, are likely to be prototypic of a selective downregulation strategy for many genes, including a subset of NF-kappaB target genes.


Assuntos
Acetiltransferases/metabolismo , Antígenos CD/genética , Proteínas do Citoesqueleto/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Glicoproteínas de Membrana/genética , Metástase Neoplásica/genética , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas/genética , Transativadores/metabolismo , Transcrição Gênica/genética , Acetiltransferases/genética , Animais , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina , Colágeno , Regulação para Baixo/genética , Combinação de Medicamentos , Histona Acetiltransferases , Humanos , Proteína Kangai-1 , Laminina , Lisina Acetiltransferase 5 , Masculino , Camundongos , NF-kappa B/metabolismo , Transplante de Neoplasias , Regiões Promotoras Genéticas/genética , Neoplasias da Próstata/metabolismo , Proteoglicanas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , beta Catenina
19.
J Biol Chem ; 284(48): 33475-84, 2009 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-19801685

RESUMO

HslVU is a bacterial ATP-dependent protease distantly related to eukaryotic proteasomes consisting of hexameric HslU ATPase and dodecameric HslV protease. As a homolog of the 20 S proteasome beta-subunits, HslV also uses the N-terminal threonine as the active site residue. However, unlike the proteasome that has only 6 active sites among the 14 beta-subunits, HslV has 12 active sites that could potentially contribute to proteolytic activity. Here, by using a series of HslV dodecamers containing different numbers of active sites, we demonstrate that like the proteasome, HslV with only approximately 6 active sites is sufficient to support full catalytic activity. However, a further reduction of the number of active sites leads to a proportional decrease in activity. Using proteasome inhibitors, we also demonstrate that substrate-mediated stabilization of the HslV-HslU interaction remains unchanged until the number of the active sites is decreased to approximately 6 but is gradually compromised upon further reduction. These results with a mathematical model suggest HslVU utilizes no more than 6 active sites at any given time, presumably because of the action of HslU. These results also suggest that each ATP-bound HslU subunit activates one HslV subunit and that substrate bound to the HslV active site stimulates the HslU ATPase activity by stabilizing the HslV-HslU interaction. We propose this mechanism plays an important role in supporting complete degradation of substrates while preventing wasteful ATP hydrolysis in the resting state by controlling the interaction between HslV and HslU through the catalytic engagement of the proteolytic active sites.


Assuntos
Trifosfato de Adenosina/metabolismo , Endopeptidase Clp/metabolismo , Proteínas de Escherichia coli/metabolismo , Treonina/metabolismo , Trifosfato de Adenosina/análogos & derivados , Alanina/genética , Alanina/metabolismo , Substituição de Aminoácidos , Sítios de Ligação , Catálise , Eletroforese em Gel de Poliacrilamida , Endopeptidase Clp/genética , Ensaios Enzimáticos , Proteínas de Escherichia coli/genética , Hidrólise , Cinética , Modelos Biológicos , Mutação , Peptídeos/metabolismo , Ligação Proteica , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Espectrometria de Fluorescência , Especificidade por Substrato , Treonina/genética
20.
Proc Natl Acad Sci U S A ; 104(52): 20793-8, 2007 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18087039

RESUMO

Posttranslational modification by small ubiquitin-like modifier (SUMO) controls diverse cellular functions of transcription factors and coregulators and participates in various cellular processes including signal transduction and transcriptional regulation. Here, we report that pontin, a component of chromatin-remodeling complexes, is SUMO-modified, and that SUMOylation of pontin is an active control mechanism for the transcriptional regulation of pontin on androgen-receptor target genes in prostate cancer cells. Biochemical purification of pontin-containing complexes revealed the presence of the Ubc9 SUMO-conjugating enzyme that underlies its function as an activator. Intriguingly, 5alpha-dihydroxytestosterone treatments significantly increased the SUMOylation of pontin, and SUMOylated pontin showed further activation of a subset of nuclear receptor-dependent transcription and led to an increase in proliferation and growth of prostate cancer cells. These data clearly define a functional model and provide a link between SUMO modification and prostate cancer progression.


Assuntos
Proteínas de Transporte/química , Cromatina/química , DNA Helicases/química , Neoplasias da Próstata/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/química , ATPases Associadas a Diversas Atividades Celulares , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Humanos , Hidroxitestosteronas/farmacologia , Lisina/química , Masculino , Modelos Biológicos , Proteína SUMO-1 , Transdução de Sinais , Transcrição Gênica , Enzimas de Conjugação de Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA