Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
5.
Mol Cell ; 31(6): 835-49, 2008 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-18922467

RESUMO

SRC-3/AIB1 is a master growth coactivator and oncogene, and phosphorylation activates it into a powerful coregulator. Dephosphorylation is a potential regulatory mechanism for SRC-3 function, but the identity of such phosphatases remains unexplored. Herein, we report that, using functional genomic screening of human Ser/Thr phosphatases targeting SRC-3's known phosphorylation sites, the phosphatases PDXP, PP1, and PP2A were identified to be key negative regulators of SRC-3 transcriptional coregulatory activity in steroid receptor signalings. PDXP and PP2A dephosphorylate SRC-3 and inhibit its ligand-dependent association with estrogen receptor. PP1 stabilizes SRC-3 protein by blocking its proteasome-dependent turnover through dephosphorylation of two previously unidentified phosphorylation sites (Ser101 and S102) required for activity. These two sites are located within a degron of SRC-3 and are primary determinants of SRC-3 turnover. Moreover, PP1 regulates the oncogenic cell proliferation and invasion functions of SRC-3 in breast cancer cells.


Assuntos
Fosfoproteínas/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Regulação da Expressão Gênica , Genoma/genética , Células HeLa , Humanos , Coativador 3 de Receptor Nuclear , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Fosfosserina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteína Fosfatase 1/metabolismo , Proteína Fosfatase 2/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Termodinâmica , Fatores de Transcrição/genética , Transcrição Gênica
6.
J Cell Sci ; 126(Pt 6): 1440-53, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23418360

RESUMO

MicroRNAs are known to play regulatory roles in gene expression associated with cancer development. We analyzed levels of the microRNA miR-24 in patients with breast carcinoma and found that miR-24 was higher in breast carcinoma samples than in benign breast tissues. We generated constructs expressing miR-24 and studied its functions using both in vitro and in vivo techniques. We found that the ectopic expression of miR-24 promoted breast cancer cell invasion and migration. In vivo experiments in mice indicated that the expression of miR-24 enhanced tumor growth, invasion into local tissues, metastasis to lung tissues and decreased overall mouse survival. In the miR-24-expressing cells and tumors, EGFR was highly phosphorylated, whereas expression of the phosphatases tyrosine-protein phosphatase non-receptor type 9 (PTPN9) and receptor-type tyrosine-protein phosphatase F (PTPRF) were repressed. We confirmed that miR-24 could directly target both PTPN9 and PTPRF. Consistent with this, we found that the levels of phosphorylated epidermal growth factor receptor (pEGFR) were higher whereas the levels of PTPN9 and PTPRF were lower in the patients with metastatic breast carcinoma. Ectopic expression of PTPN9 and PTPRF decreased pEGFR levels, cell invasion, migration and tumor metastasis. Furthermore, we found that MMP2, MMP11, pErk, and ADAM15 were upregulated, whereas TIMP2 was downregulated; all of which supported the roles of miR-24 in tumor invasion and metastasis. Our results suggest that miR-24 plays a key role in breast cancer invasion and metastasis. miR-24 could potentially be a target for cancer intervention.


Assuntos
Neoplasias da Mama/patologia , MicroRNAs/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Animais , Neoplasias da Mama/genética , Processos de Crescimento Celular/genética , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , Invasividade Neoplásica/genética , Metástase Neoplásica , Transplante de Neoplasias , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/genética , Transdução de Sinais/genética , Transgenes/genética
7.
Theranostics ; 14(6): 2345-2366, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646645

RESUMO

Rationale: Primordial follicles are limited in number and cannot be regenerated, dormant primordial follicles cannot be reversed once they enter a growth state. Therefore, the length of the female reproductive lifespan depends on the orderly progression and selective activation of primordial follicles, the mechanism of which remains unclear. Methods: We used human ovarian cortical biopsy specimens, granulosa cells from diminished ovarian reserve (DOR) patients, Hdac6-overexpressing transgenic mouse model, and RNA sequencing to analyze the crucial roles of histone deacetylase 6 (HDAC6) in fertility preservation and primordial follicle activation. Results: In the present study, we found that HDAC6 was highly expressed in most dormant primordial follicles. The HDAC6 expression was reduced accompanying reproductive senescence in human and mouse ovaries. Overexpression of Hdac6 delayed the rate of primordial follicle activation, thereby prolonging the mouse reproductive lifespan. Short-term inhibition of HDAC6 promoted primordial follicle activation and follicular development in humans and mice. Mechanism studies revealed that HDAC6 directly interacted with NGF, reducing acetylation modification of NGF and thereby accelerating its ubiquitination degradation. Consequently, the reduced NGF protein level maintained the dormancy of primordial follicles. Conclusions: The physiological significance of the high expression of HDAC6 in most primordial follicles is to reduce NGF expression and prevent primordial follicle activation to maintain female fertility. Reduced HDAC6 expression increases NGF expression in primordial follicles, activating their development and contributing to reproduction. Our study provides a clinical reference value for fertility preservation.


Assuntos
Desacetilase 6 de Histona , Camundongos Transgênicos , Fator de Crescimento Neural , Folículo Ovariano , Ubiquitinação , Animais , Feminino , Humanos , Camundongos , Acetilação , Células da Granulosa/metabolismo , Desacetilase 6 de Histona/metabolismo , Desacetilase 6 de Histona/genética , Fator de Crescimento Neural/metabolismo , Folículo Ovariano/metabolismo
8.
Dev Cell ; 11(3): 301-12, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16950122

RESUMO

To assess whether Smad signaling affects skin development, we generated transgenic mice in which a Smad antagonist, Smad7, was induced in keratinocytes, including epidermal stem cells. Smad7 transgene induction perturbed hair follicle morphogenesis and differentiation, but accelerated sebaceous gland morphogenesis. Further analysis revealed that independent of its role in anti-Smad signaling, Smad7 bound beta-catenin and induced beta-catenin degradation by recruiting an E3 ligase, Smurf2, to the Smad7/beta-catenin complex. Consequently, Wnt/beta-catenin signaling was suppressed in Smad7 transgenic hair follicles. Coexpression of the Smurf2 and Smad7 transgenes exacerbated Smad7-induced abnormalities in hair follicles and sebaceous glands. Conversely, when endogenous Smad7 was knocked down, keratinocytes exhibited increased beta-catenin protein and enhanced Wnt signaling. Our data reveal a mechanism for Smad7 in antagonizing Wnt/beta-catenin signaling, thereby shifting the skin differentiation program from forming hair follicles to sebaceous glands.


Assuntos
Epiderme/patologia , Folículo Piloso/fisiologia , Proteína Smad7/fisiologia , Células-Tronco/fisiologia , beta Catenina/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Regulação para Baixo , Epiderme/metabolismo , Folículo Piloso/metabolismo , Humanos , Queratinócitos/metabolismo , Camundongos , Camundongos Transgênicos , Morfogênese , Glândulas Sebáceas/fisiologia , Transdução de Sinais , Proteína Smad7/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Wnt/metabolismo
9.
Chemosphere ; 278: 130392, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33819894

RESUMO

The reaction process of petrochemical sludge under hydrothermal conditions was investigated to explore the feasibility of its disposal using hydrothermal treatment. Experiments were conducted in an autoclave for 30 min at 350-450 °C. During the hydrothermal treat of petrochemical sludge, 44.98%-59.64% of the oil (organic matter) in the sludge was decomposed into aqueous and gas products. The gas yield reached 1.37 mol/kg of organic matter at reaction temperature of 450 °C. The H2 yield was 0.26 mol/kg of organic matter. The primary product was still in the aqueous phase. The TOC concentration of the aqueous product was in the range of 14,960-19,050 mg/L. The concentration of COD, total phenol, and total nitrogen of product were in the ranges of 9029-10,870, 13.83-20.10, and 497.5-599.0 mg/L, respectively. The group analysis and GC-MS analysis of the residual oil indicated that the saturated long chain hydrocarbons (C18-C21) in petrochemical sludge had decomposed to form saturated short chain hydrocarbons (C11-C17); however, the short chain saturated hydrocarbons in the sludge had decomposed thoroughly. The removal rate of asphaltenes, resins and aromatic hydrocarbons were low. Finally, a mechanism for treating petrochemical sludge under hydrothermal conditions was proposed. The study provides an experimental basis for the hydrothermal treatment of petrochemical sludge.


Assuntos
Esgotos , Água , Hidrocarbonetos , Nitrogênio , Temperatura
10.
Mol Cell Biol ; 24(17): 7524-37, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15314162

RESUMO

Smad4/DPC4, a common signal transducer in transforming growth factor beta (TGF-beta) signaling, is frequently inactivated in human cancer. Although the ubiquitin-proteasome pathway has been established as one mechanism of inactivating Smad4 in cancer, the specific ubiquitin E3 ligase for ubiquitination-mediated proteolysis of Smad4 cancer mutants remains unclear. In this report, we identified the SCFSkp2 complex as candidate Smad4-interacting proteins in an antibody array-based screen and further elucidated the functions of SCFSkp2 in mediating the metabolic instability of cancer-derived Smad4 mutants. We found that Skp2, the F-box component of SCFSkp2, physically interacted with Smad4 at the physiological levels. Several cancer-derived unstable mutants exhibited significantly increased binding to Skp2, which led to their increased ubiquitination and accelerated proteolysis. These results suggest an important role for the SCFSkp2 complex in switching cancer mutants of Smad4 to undergo polyubiquitination-dependent degradation.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Neoplasias/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Transativadores/genética , Transativadores/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Motivos de Aminoácidos , Animais , Linhagem Celular , Cisteína Endopeptidases/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Complexos Multienzimáticos/metabolismo , Neoplasias/genética , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Processamento de Proteína Pós-Traducional , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Transdução de Sinais/fisiologia , Proteína Smad4 , Ubiquitina-Proteína Ligases/genética , Proteínas Quinases p38 Ativadas por Mitógeno
11.
Mol Cell Biol ; 23(24): 9081-93, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14645520

RESUMO

Smad6 and Smad7 are inhibitory Smads induced by transforming growth factor beta-Smad signal transduction pathways in a negative-feedback mechanism. Previously it has been thought that inhibitory Smads bind to the type I receptor and block the phosphorylation of receptor-activated Smads, thereby inhibiting the initiation of Smad signaling. Conversely, few studies have suggested the possible nuclear functions of inhibitory Smads. Here, we present compelling evidence demonstrating that Smad6 repressed bone morphogenetic protein-induced Id1 transcription through recruiting transcriptional corepressor C-terminal binding protein (CtBP). A consensus CtBP-binding motif, PLDLS, was identified in the linker region of Smad6. Our findings show that mutation in the motif abolished the Smad6 binding to CtBP and subsequently its repressor activity of transcription. We conclude that the nuclear functions and physical interaction of Smad6 and CtBP provide a novel mechanism for the transcriptional regulation by inhibitory Smads.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fosfoproteínas/metabolismo , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta , Oxirredutases do Álcool , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/genética , Linhagem Celular , DNA/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Células HeLa , Humanos , Proteína 1 Inibidora de Diferenciação , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Proteína Smad6 , Proteína Smad7 , Transativadores/química , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
12.
Am J Clin Exp Urol ; 2(3): 239-48, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25374926

RESUMO

Transforming Growth Factor-ß (TGF-ß) regulates the reactive stroma microenvironment associated with most carcinomas and mediates expression of many stromal derived factors important for tumor progression, including FGF-2 and CTGF. TGF-ß is over-expressed in most carcinomas, and FGF-2 action is important in tumor-induced angiogenesis. The signaling mechanisms of how TGF-ß regulates FGF-2 expression in the reactive stroma microenvironment are not understood. Accordingly, we have assessed key signaling pathways that mediate TGF-ß1-induced FGF-2 expression in prostate stromal fibroblasts and mouse embryo fibroblasts (MEFs) null for Smad2 and Smad3. TGF-ß1 induced phosphorylation of Smad2, Smad3, p38 and ERK1/2 proteins in both control MEFs and prostate fibroblasts. Of these, Smad3, but not Smad2 was found to be required for TGF-ß1 induction of FGF-2 expression in stromal cells. ChIP analysis revealed a Smad3/Smad4 complex was associated with the -1.9 to -2.3 kb upstream proximal promoter of the FGF-2 gene, further suggesting a Smad3-specific regulation. In addition, chemical inhibition of p38 or ERK1/2 MAPK activity also blocked TGF-ß1-induced FGF-2 expression in a Smad3-independent manner. Conversely, inhibition of JNK signaling enhanced FGF-2 expression. Together, these data indicate that expression of FGF-2 in fibroblasts in the tumor stromal cell microenvironment is coordinately dependent on both intact Smad3 and MAP kinase signaling pathways. These pathways and key downstream mediators of TGF-ß action in the tumor reactive stroma microenvironment, may evolve as putative targets for therapeutic intervention.

13.
Artigo em Inglês | MEDLINE | ID: mdl-22645523

RESUMO

Nodal is a member of the transforming growth factor-ß (TGF-ß) superfamily that plays critical roles during embryogenesis. Recent studies in ovarian, breast, prostate, and skin cancer cells suggest that Nodal also regulates cell proliferation, apoptosis, and invasion in cancer cells. However, it appears to exert both tumor-suppressing and tumor-promoting effects, depending on the cell type. To further understand the role of Nodal in tumorigenesis, we examined the effect of Nodal in glioblastoma cell growth and spheroid formation using U87 cell line. Treatment of U87 with recombinant Nodal significantly increased U87 cell growth. In U87 cells stably transfected with the plasmid encoding Nodal, Smad2 phosphorylation was strongly induced and cell growth was significantly enhanced. Overexpression of Nodal also resulted in tight spheroid formation. On the other hand, the cells stably transfected with Nodal siRNA formed loose spheroids. Nodal is known to signal through activin receptor-like kinase 4 (ALK4) and ALK7 and the Smad2/3 pathway. To determine which receptor and Smad mediate the growth promoting effect of Nodal, we transfected siRNAs targeting ALK4, ALK7, Smad2, or Smad3 into Nodal-overexpressing cells and observed that cell growth was significantly inhibited by ALK4, ALK7, and Smad3 siRNAs. Taken together, these findings suggest that Nodal may have tumor-promoting effects on glioblastoma cells and these effects are mediated by ALK4, ALK7, and Smad3.

14.
Methods Mol Biol ; 647: 125-37, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20694664

RESUMO

In eukaryotes, regulation of signaling mediators/effectors in the nucleus is one of the principal mechanisms that govern duration and strength of signaling. Smads are a family of structurally related intracellular proteins that serve as signaling effectors for transforming growth factor beta (TGF-beta) and TGF-beta-related proteins. Accumulating evidence demonstrates that Smads possess intrinsic nucleocytoplasmic shuttling capacity, which enables them to transmit TGF-beta signals from cell membrane to nucleus. We recently identified two important steps in the termination of nuclear Smad signaling. The first step is initiated by a serine/threonine phosphatase PPM1A that dephosphorylates Smad2/3 in the nucleus, thereby shutting down signaling capacity of phosphorylated Smad2/3. The second step involves nuclear export of dephosphorylated Smad2/3 with the aid of nuclear protein RanBP3 to terminate Smad signaling. This chapter introduces methods for examining nuclear export of Smad2/3 in TGF-beta signaling.


Assuntos
Núcleo Celular/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Transporte Ativo do Núcleo Celular , Fracionamento Celular , Linhagem Celular , Humanos , Espaço Intracelular/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação
15.
Cell Res ; 19(1): 140-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19079362

RESUMO

Id1 is a member of the inhibitor of differentiation (Id) protein family that regulates a wide range of cell functions. Previous studies have shown that expression of the Id1 gene is down-regulated by TGF-beta in epithelial cells, whereas it is up-regulated by BMP in a variety of cell types. During our study of the biological function of TGF-beta1, we found that Id1 can be strongly up-regulated by TGF-beta1 in the human mammary gland epithelial cell line MCF10A. Quantitative real-time RT-PCR has revealed as high as 7.5-fold induction of Id1 mRNA by TGF-beta1 in MCF10A cells after 1 h of TGF-beta1 stimulation, and this induction does not require de novo protein synthesis. Using Smad knockdown and knockout approaches, we have identified Smad3 as the responsible R-Smad for mediating transcriptional activation of the Id1 gene. Chromatin immunoprecipitation assay confirms that Smad3 and Smad4 bind to the upstream region of the Id1 gene. Our results demonstrate that Smad3, but not Smad2, mediates TGF-beta1-dependent early transcriptional induction of Id1.


Assuntos
Proteína 1 Inibidora de Diferenciação/biossíntese , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Acetilação , Animais , Cromatina/imunologia , Células Epiteliais , Técnicas de Silenciamento de Genes , Histonas/metabolismo , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Camundongos , Regiões Promotoras Genéticas , Transdução de Sinais , Proteína Smad3/imunologia , Fatores de Transcrição
16.
J Biol Chem ; 283(48): 33578-84, 2008 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-18829461

RESUMO

Cdk9 is the catalytic subunit of a general RNA polymerase II elongation factor known as positive transcription elongation factor b (P-TEFb). The kinase function of P-TEFb requires phosphorylation of Thr-186 in the T-loop of Cdk9 to allow substrates to access the catalytic core of the enzyme. To identify human phosphatases that dephosphorylate the T-loop of Cdk9, we used a Thr-186-phosphospecific antiserum to screen a phosphatase expression library. Overexpression of PPM1A and the related PPM1B greatly reduced Cdk9 T-loop phosphorylation in vivo. PPM1A and Cdk9 appear to associate in vivo as the proteins could be co-immunoprecipitated. The short hairpin RNA depletion of PPM1A resulted in an increase in Cdk9 T-loop phosphorylation. In phosphatase reactions in vitro, purified PPM1A could dephosphorylate Thr-186 both with and without the association of 7SK RNA, a small nuclear RNA that is bound to approximately 50% of total cellular P-TEFb. PPM1B only efficiently dephosphorylated Cdk9 Thr-186 in vitro when 7SK RNA was depleted from P-TEFb. Taken together, our data indicate that PPM1A and to some extent PPM1B are important negative regulators of P-TEFb function.


Assuntos
Quinase 9 Dependente de Ciclina/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fator B de Elongação Transcricional Positiva/metabolismo , Domínio Catalítico/fisiologia , Quinase 9 Dependente de Ciclina/genética , Células HeLa , Humanos , Fosfoproteínas Fosfatases/genética , Fosforilação/fisiologia , Fator B de Elongação Transcricional Positiva/genética , Proteína Fosfatase 2C , Estrutura Secundária de Proteína/fisiologia , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/metabolismo
17.
J Biol Chem ; 281(48): 36526-32, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16931515

RESUMO

Bone morphogenetic proteins (BMPs) are secreted polypeptides belonging to the transforming growth factor-beta (TGF-beta) superfamily that activates a broad range of biological responses in the metazoan organism. The BMP-initiated signaling pathway is under tight control by processes including regulation of the ligands, the receptors, and the key downstream intracellular effector Smads. A critical point of control in BMP signaling is the phosphorylation of Smad1, Smad5, and Smad8 in their C-terminal SXS motif. Although such phosphorylation, which is mediated by the type I BMP receptor kinases in response to BMP stimulation, is well characterized, biochemical mechanisms underlying Smad dephosphorylation remain to be elucidated. In this study, we have found that PPM1A, a metal ion-dependent protein serine/threonine phosphatase, physically interacts with and dephosphorylates Smad1 both in vitro and in vivo. Functionally, overexpression of PPM1A abolishes BMP-induced transcriptional responses, whereas RNA interference-mediated knockdown of PPM1A enhances BMP signaling. Collectively, our study suggests that PPM1A plays an important role in controlling BMP signaling through catalyzing Smad dephosphorylation.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Fosfoproteínas Fosfatases/química , Proteína Smad1/fisiologia , Motivos de Aminoácidos , Animais , Western Blotting , Catálise , Linhagem Celular , Humanos , Imunoprecipitação , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Mutação Puntual , Proteína Fosfatase 2C , Transdução de Sinais , Proteína Smad1/metabolismo , Transfecção , Fator de Crescimento Transformador beta/metabolismo
18.
Cell ; 125(5): 915-28, 2006 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-16751101

RESUMO

TGFbeta signaling controls diverse normal developmental processes and pathogenesis of diseases including cancer and autoimmune and fibrotic diseases. TGFbeta responses are generally mediated through transcriptional functions of Smads. A key step in TGFbeta signaling is ligand-induced phosphorylation of receptor-activated Smads (R-Smads) catalyzed by the TGFbeta type I receptor kinase. However, the potential of Smad dephosphorylation as a regulatory mechanism of TGFbeta signaling and the identity of Smad-specific phosphatases remain elusive. Using a functional genomic approach, we have identified PPM1A/PP2Calpha as a bona fide Smad phosphatase. PPM1A dephosphorylates and promotes nuclear export of TGFbeta-activated Smad2/3. Ectopic expression of PPM1A abolishes TGFbeta-induced antiproliferative and transcriptional responses, whereas depletion of PPM1A enhances TGFbeta signaling in mammalian cells. Smad-antagonizing activity of PPM1A is also observed during Nodal-dependent early embryogenesis in zebrafish. This work demonstrates that PPM1A/PP2Calpha, through dephosphorylation of Smad2/3, plays a critical role in terminating TGFbeta signaling.


Assuntos
Fosfoproteínas Fosfatases/metabolismo , Transdução de Sinais/fisiologia , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Receptores de Ativinas Tipo I/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Embrião não Mamífero , Humanos , Fosfoproteínas Fosfatases/genética , Fosforilação , Proteína Fosfatase 2C , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Regulação para Cima/fisiologia , Peixe-Zebra
19.
J Biol Chem ; 280(25): 24227-37, 2005 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-15849193

RESUMO

Smads, key effectors of transforming growth factor (TGF)-beta, activin, and bone morphogenetic protein (BMP) signaling, regulate gene expression and interact with coactivators and corepressors that modulate Smad activity. The corepressor Evi-1 exerts its oncogenic effects by repressing TGF-beta/Smad3-mediated transcription, thereby blocking TGF-beta-induced growth arrest. Because Evi-1 interacts with the highly conserved MH2 domain of Smad3, we investigated the physical and functional interaction of Evi-1 with Smad1 and Smad2, downstream targets of BMP and activin signaling, respectively. Evi-1 interacted with and repressed the receptor-activated transcription through Smad1 and Smad2, similarly to Smad3. In addition, Evi-1 repressed BMP/Smad1- and activin/Smad2-mediated induction of endogenous Xenopus gene expression, suggesting a role of repression of BMP and activin signals by Evi-1 in vertebrate embryogenesis. Evi-1 also repressed the induction of endogenous Smad7 expression by TGF-beta family ligands. In the course of these studies, we observed Evi-1 repression of Smad transactivation even when Smad binding to DNA was kept constant. We therefore explored the mechanism of Evi-1 repression of TGF-beta family-inducible transcription. Evi-1 repression did not result from displacement of Smad binding to DNA or to CREB-binding protein but from the recruitment of Evi-1 by Smad3 and CREB-binding protein to DNA. Following TGF-beta stimulation, Evi-1 and the associated corepressor CtBP were recruited to the endogenous Smad7 promoter. Evi-1 recruitment to the promoter decreased TGF-beta-induced histone acetylation, coincident with its repression of Smad7 gene expression. In this way, Evi-1 acts as a general Smad corepressor to inhibit TGF-beta-, activin-, and BMP-inducible transcription.


Assuntos
Ativinas/fisiologia , Proteínas Morfogenéticas Ósseas/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proto-Oncogenes/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Acetilação , Animais , Proteína de Ligação a CREB , Linhagem Celular , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histonas/metabolismo , Humanos , Proteína do Locus do Complexo MDS1 e EVI1 , Proteínas Nucleares/fisiologia , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Ligação Proteica , Proteína Smad3 , Transativadores/fisiologia , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Xenopus
20.
J Biol Chem ; 278(33): 31043-8, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12813045

RESUMO

Tumor suppressor Smad4/DPC4 is a central intracellular signal transducer for transforming growth factor-beta (TGF-beta) signaling. We recently reported that transcriptional potential of Smad4 was regulated by SUMOylation in transfected HeLa cells (1), but the precise mechanism and function of Smad4 SUMOylation in TGF-beta signaling remain to be elucidated. Here, we describe the regulation of TGF-beta signaling by SUMOylation through the control of Smad4 metabolic stability and subcellular localization. We found that SUMO-1 overexpression strongly increases Smad4 levels, while inhibition of SUMOylation by small interfering RNA (siRNA)-mediated knockdown of the E2 enzyme Ubc9 reduces endogenous Smad4 levels. Concomitantly, SUMO-1 overexpression enhances and Ubc9 knockdown reduces levels of intranuclear Smad4, growth inhibitory response, as well as transcriptional responses to TGF-beta. Comparison of wild type and mutant forms of Smad4 for SUMOylation, ubiquitination, and half-life allows the conclusion that SUMO-1 modification serves to protect Smad4 from ubiquitin-dependent degradation and consequently enhances the growth inhibitory and transcriptional responses of Smad4.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteína SUMO-1/metabolismo , Transativadores/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Divisão Celular/fisiologia , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Células HeLa , Humanos , Mutação , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , RNA Interferente Pequeno , Transdução de Sinais/fisiologia , Proteína Smad4 , Transativadores/genética , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Proteínas Supressoras de Tumor/genética , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa