Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 288(34): 24636-46, 2013 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-23864657

RESUMO

The functional significance of the widespread down-regulation of Rap1 GTPase-activating protein (Rap1GAP), a negative regulator of Rap activity, in human tumors is unknown. Here we show that human colon cancer cells depleted of Rap1GAP are endowed with more aggressive migratory and invasive properties. Silencing Rap1GAP enhanced the migration of confluent and single cells. In the latter, migration distance, velocity, and directionality were increased. Enhanced migration was a consequence of increased endogenous Rap activity as silencing Rap expression selectively abolished the migration of Rap1GAP-depleted cells. ROCK-mediated cell contractility was suppressed in Rap1GAP-depleted cells, which exhibited a spindle-shaped morphology and abundant membrane protrusions. Tumor cells can switch between Rho/ROCK-mediated contractility-based migration and Rac1-mediated mesenchymal motility. Strikingly, the migration of Rap1GAP-depleted, but not control cells required Rac1 activity, suggesting that loss of Rap1GAP alters migratory mechanisms. Inhibition of Rac1 activity restored membrane blebbing and increased ROCK activity in Rap1GAP-depleted cells, suggesting that Rac1 contributes to the suppression of contractility. Collectively, these findings identify Rap1GAP as a critical regulator of aggressive tumor cell behavior and suggest that the level of Rap1GAP expression influences the migratory mechanisms that are operative in tumor cells.


Assuntos
Movimento Celular , Proteínas Ativadoras de GTPase/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Linhagem Celular Tumoral , Proteínas Ativadoras de GTPase/genética , Humanos , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
2.
Endocr Relat Cancer ; 19(4): 575-88, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22696507

RESUMO

The functional significance of decreased RAP1GAP protein expression in human tumors is unclear. To identify targets of RAP1GAP downregulation in the thyroid gland, RAP1 and RAP2 protein expression in human thyroid cells and in primary thyroid tumors were analyzed. RAP1GAP and RAP2 were co-expressed in normal thyroid follicular cells. Intriguingly, RAP1 was not detected in normal thyroid cells, although it was detected in papillary thyroid carcinomas, which also expressed RAP2. Both RAP proteins were detected at the membrane in papillary thyroid tumors, suggesting that they are activated when RAP1GAP is downregulated. To explore the functional significance of RAP1GAP depletion, RAP1GAP was transiently expressed at the lowest level that is sufficient to block endogenous RAP2 activity in papillary and anaplastic thyroid carcinoma cell lines. RAP1GAP impaired the ability of cells to spread and migrate on collagen. Although RAP1GAP had no effect on protein tyrosine phosphorylation in growing cells, RAP1GAP impaired phosphorylation of focal adhesion kinase and paxillin at sites phosphorylated by SRC in cells acutely plated on collagen. SRC activity was increased in suspended cells, where it was inhibited by RAP1GAP. Inhibition of SRC kinase activity impaired cell spreading and motility. These findings identify SRC as a target of RAP1GAP depletion and suggest that the downregulation of RAP1GAP in thyroid tumors enhances SRC-dependent signals that regulate cellular architecture and motility.


Assuntos
Carcinoma Papilar/patologia , Movimento Celular , Citoesqueleto/fisiologia , Proteínas Ativadoras de GTPase/fisiologia , Neoplasias da Glândula Tireoide/patologia , Carcinoma Papilar/genética , Carcinoma Papilar/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/fisiologia , Citoesqueleto/metabolismo , Regulação para Baixo/genética , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HT29 , Humanos , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Carcinoma Anaplásico da Tireoide , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/metabolismo , Transfecção , Proteínas rap de Ligação ao GTP/genética , Proteínas rap de Ligação ao GTP/metabolismo , Proteínas rap1 de Ligação ao GTP/genética , Proteínas rap1 de Ligação ao GTP/metabolismo
3.
Cell Adh Migr ; 5(4): 323-31, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21785277

RESUMO

The significance of the widespread downregulation of Rap1GAP in human tumors is unknown. In previous studies we demonstrated that silencing Rap1GAP expression in human colon cancer cells resulted in sustained increases in Rap activity, enhanced spreading on collagen and the weakening of cell-cell contacts. The latter finding was unexpected based on the role of Rap1 in strengthening cell-cell adhesion and reports that Rap1GAP impairs cell-cell adhesion. We now show that Rap1GAP is a more effective inhibitor of cell-matrix compared to cell-cell adhesion. Overexpression of Rap1GAP in human colon cancer cells impaired Rap2 activity and the ability of cells to spread and migrate on collagen IV. Under the same conditions, Rap1GAP had no effect on cell-cell adhesion. Overexpression of Rap1GAP did not enhance the dissociation of cell aggregates nor did it impair the accumulation of ß-catenin and E-cadherin at cell-cell contacts. To further explore the role of Rap1GAP in the regulation of cell-cell adhesion, Rap1GAP was overexpressed in non-transformed thyroid epithelial cells. Although the formation of cell-cell contacts required Rap1, overexpression of Rap1GAP did not impair cell-cell adhesion. These data indicate that transient, modest expression of Rap1GAP is compatible with cell-cell adhesion and that the role of Rap1GAP in the regulation of cell-cell adhesion may be more complex than is currently appreciated.


Assuntos
Junções Célula-Matriz/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Regulação Neoplásica da Expressão Gênica , Adenoviridae/genética , Animais , Antígenos CD , Caderinas/metabolismo , Adesão Celular , Movimento Celular , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Células Epiteliais/metabolismo , Células HCT116 , Humanos , Interferência de RNA , Ratos , Ratos Wistar , Transdução de Sinais , Transfecção , beta Catenina/metabolismo , Proteínas rap de Ligação ao GTP/metabolismo
4.
Endocr Relat Cancer ; 18(3): 301-10, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21367844

RESUMO

Increases in Rap activity have been associated with tumor progression. Although activating mutations in Rap have not been described, downregulation of Rap1GAP is frequent in human tumors including thyroid carcinomas. In this study, we explored whether endogenous Rap1GAP expression could be restored to thyroid tumor cells. The effects of deacetylase inhibitors and a demethylating agent, individually and in combination, were examined in four differentiated and six anaplastic thyroid carcinoma (ATC) cell lines. Treatment with the structurally distinct histone deacetylase (HDAC) inhibitors, sodium butyrate and trichostatin A, increased Rap1GAP expression in all the differentiated thyroid carcinoma cell lines and in four of the six ATC cell lines. The demethylating agent, 5-aza-deoxycytidine, restored Rap1GAP expression in one anaplastic cell line and enhanced the effects of HDAC inhibitors in a second anaplastic cell line. Western blotting indicated that Rap2 was highly expressed in human thyroid cancer cells. Importantly, treatment with HDAC inhibitors impaired Rap2 activity in both differentiated and anaplastic tumor cell lines. The mechanism through which Rap activity is repressed appears to entail effects on the expression of multiple Rap regulators, including RapGEFs and RapGAPs. These results suggest that HDAC inhibitors may provide a tractable approach to impair Rap activity in human tumor cells.


Assuntos
Butiratos/farmacologia , Carcinoma/patologia , Proteínas Ativadoras de GTPase/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Proteínas de Neoplasias/biossíntese , Neoplasias da Glândula Tireoide/patologia , Proteínas rap de Ligação ao GTP/antagonistas & inibidores , Proteínas rap1 de Ligação ao GTP/antagonistas & inibidores , Adenocarcinoma Folicular/metabolismo , Adenocarcinoma Folicular/patologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Carcinoma/metabolismo , Carcinoma Papilar/metabolismo , Carcinoma Papilar/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Metilação de DNA/efeitos dos fármacos , Decitabina , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Proteínas Ativadoras de GTPase/genética , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , RNA Interferente Pequeno/farmacologia , Neoplasias da Glândula Tireoide/metabolismo , Regulação para Cima/efeitos dos fármacos
5.
Mol Cell Biol ; 30(13): 3262-74, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20439492

RESUMO

Rap1GAP expression is decreased in human tumors. The significance of its downregulation is unknown. We show that Rap1GAP expression is decreased in primary colorectal carcinomas. To elucidate the advantages conferred on tumor cells by loss of Rap1GAP, Rap1GAP expression was silenced in human colon carcinoma cells. Suppressing Rap1GAP induced profound alterations in cell adhesion. Rap1GAP-depleted cells exhibited defects in cell/cell adhesion that included an aberrant distribution of adherens junction proteins. Depletion of Rap1GAP enhanced adhesion and spreading on collagen. Silencing of Rap expression normalized spreading and restored E-cadherin, beta-catenin, and p120-catenin to cell/cell contacts, indicating that unrestrained Rap activity underlies the alterations in cell adhesion. The defects in adherens junction protein distribution required integrin signaling as E-cadherin and p120-catenin were restored at cell/cell contacts when cells were plated on poly-l-lysine. Unexpectedly, Src activity was increased in Rap1GAP-depleted cells. Inhibition of Src impaired spreading and restored E-cadherin at cell/cell contacts. These findings provide the first evidence that Rap1GAP contributes to cell/cell adhesion and highlight a role for Rap1GAP in regulating cell/matrix and cell/cell adhesion. The frequent downregulation of Rap1GAP in epithelial tumors where alterations in cell/cell and cell/matrix adhesion are early steps in tumor dissemination supports a role for Rap1GAP depletion in tumor progression.


Assuntos
Adesão Celular/fisiologia , Linhagem Celular Tumoral/metabolismo , Neoplasias Colorretais/metabolismo , Matriz Extracelular/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Junções Aderentes/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Proteínas Ativadoras de GTPase/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Quinases da Família src/genética , Quinases da Família src/metabolismo
6.
Mol Pharmacol ; 76(5): 1123-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19720729

RESUMO

Thyrotropin (TSH) regulates thyroid cell proliferation and function through cAMP-mediated signaling pathways that activate protein kinase A (PKA) and Epac/Rap1. The respective roles of PKA versus Epac/Rap1 in TSH signaling remain unclear. We set out to determine whether PKA and/or Rap1 mediate extracellular signal-regulated kinase (ERK) activation by TSH. Neither blocking Rap1 activity nor silencing the expression of Rap1 impaired TSH or forskolin-induced ERK activation in Wistar rat thyroid cells. Direct activation of Epac1 failed to stimulate ERK activity in starved cells, suggesting that Epac-induced Rap1 activity is not coupled to ERK activation in rat thyroid cells. By contrast, PKA activity was required for cAMP-stimulated ERK phosphorylation and was sufficient to increase ERK phosphorylation in starved cells. Expression of dominant-negative Ras inhibited ERK activation by TSH, forskolin, and N(6)-monobutyryl (6MB)-cAMP, a selective activator of PKA. Silencing the expression of B-Raf also inhibited ERK activation by TSH, forskolin, and 6MB-cAMP, but not that stimulated by insulin or serum. Depletion of B-Raf impaired TSH-induced DNA synthesis, indicating a functional role for B-Raf in TSH-regulated proliferation. Collectively, these results position PKA, Ras, and B-Raf as upstream regulators of ERK activation and identify B-Raf as a selective target of cAMP-elevating agents in thyroid cells. These data provide the first evidence for a functional role for B-Raf in TSH signaling.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Proto-Oncogênicas B-raf/fisiologia , Tireotropina/fisiologia , Animais , Linhagem Celular , AMP Cíclico/fisiologia , Replicação do DNA/fisiologia , Ativação Enzimática/fisiologia , Ratos , Ratos Wistar
7.
J Clin Endocrinol Metab ; 94(3): 1026-32, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19066305

RESUMO

CONTEXT: Rap1 GTPase-activating protein (GAP) regulates the activity of Rap1, a putative oncogene. We previously reported Rap1GAP was highly expressed in normal human thyroid cells and decreased in five papillary thyroid carcinomas (PTCs). OBJECTIVES: To confirm the significance of these findings, we analyzed Rap1GAP expression in a larger set of benign tumors (adenomas and hyperplastic nodules) and PTCs. We determined whether the presence of the BRAF(V600E) mutation or allelic loss of Rap1GAP related to changes in Rap1GAP protein expression. To determine the consequences of Rap1GAP loss, we targeted Rap1GAP in culture using small interfering RNA. DESIGN, PATIENTS, AND METHODS: A highly specific Rap1GAP antibody was applied to sections of 55 human thyroid tissues. Genomic DNA was analyzed for the presence of the BRAF(V600E) mutation, and loss of Rap1GAP. Rap1GAP expression in rat thyroid cells was abolished using small interfering RNA. RESULTS: We observed that down-regulation of Rap1GAP in benign lesions and PTCs was common. Rap1GAP expression was more severely decreased in PTCs. Loss of Rap1GAP expression was observed in multiple histological variants of PTCs. Approximately 20% of PTCs and adenomas exhibited allelic loss of Rap1GAP. Loss of Rap1GAP was not associated with the presence of the BRAF(V600E) mutation. In vitro, loss of Rap1GAP was sufficient to increase Rap1 activity in thyroid cells. CONCLUSIONS: These data indicate that loss of Rap1GAP is a frequent event in PTC. The more frequent and greater down-regulation of Rap1GAP in PTCs compared with adenomas suggests a role for Rap1GAP depletion in the progression of human thyroid tumors, possibly through unrestrained Rap activity.


Assuntos
Carcinoma Papilar/química , Proteínas Ativadoras de GTPase/análise , Neoplasias da Glândula Tireoide/química , Carcinoma Papilar/genética , Carcinoma Papilar/patologia , Proteínas Ativadoras de GTPase/fisiologia , Genótipo , Humanos , Imuno-Histoquímica , Proteínas Proto-Oncogênicas B-raf/genética , Complexo Shelterina , Proteínas de Ligação a Telômeros/metabolismo , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia
8.
Virology ; 372(1): 10-23, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18006034

RESUMO

TULA, a recently identified UBA- and SH3-containing protein, has previously been shown to regulate cell signaling through protein tyrosine kinases. In order to search for novel functions of TULA, we identified, using mass spectrometry, proteins associated with TULA. ABCE-1 also known as RLI and HP68, a host factor of HIV-1 assembly, was found among TULA-associated proteins in these experiments. Considering an important role of ABCE-1 in HIV-1 assembly, we were compelled to analyze the effect of TULA on HIV-1 biogenesis. Our study provides evidence that TULA proteins substantially inhibit production of both sub-genomic and full-length HIV-1 viral particles and that the effect of TULA is dependent on UBA domain-mediated interactions. The primary role of ABCE-1 in the effect of TULA appears to be the recruitment of TULA to the sites of HIV-1 assembly where TULA interferes with the late steps of the HIV-1 life cycle, most likely by disrupting essential ubiquitylation-dependent events that remain to be identified.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Transporte/metabolismo , Chaperoninas/metabolismo , HIV-1/fisiologia , Ubiquitina/metabolismo , Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/genética , Sequência de Aminoácidos , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular , Chaperoninas/química , Chaperoninas/genética , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , HIV-1/efeitos dos fármacos , Células HeLa , Humanos , Imunoprecipitação , Espectrometria de Massas , Proteínas de Membrana , Dados de Sequência Molecular , Transfecção , Ubiquitina/genética , Montagem de Vírus
9.
Mol Cell Biol ; 27(19): 6647-58, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17646383

RESUMO

Although abundant in well-differentiated rat thyroid cells, Rap1GAP expression was extinguished in a subset of human thyroid tumor-derived cell lines. Intriguingly, Rap1GAP was downregulated selectively in tumor cell lines that had acquired a mesenchymal morphology. Restoring Rap1GAP expression to these cells inhibited cell migration and invasion, effects that were correlated with the inhibition of Rap1 and Rac1 activity. The reexpression of Rap1GAP also inhibited DNA synthesis and anchorage-independent proliferation. Conversely, eliminating Rap1GAP expression in rat thyroid cells induced a transient increase in cell number. Strikingly, Rap1GAP expression was abolished by Ras transformation. The downregulation of Rap1GAP by Ras required the activation of the Raf/MEK/extracellular signal-regulated kinase cascade and was correlated with the induction of mesenchymal morphology and migratory behavior. Remarkably, the acute expression of oncogenic Ras was sufficient to downregulate Rap1GAP expression in rat thyroid cells, identifying Rap1GAP as a novel target of oncogenic Ras. Collectively, these data implicate Rap1GAP as a putative tumor/invasion suppressor in the thyroid. In support of that notion, Rap1GAP was highly expressed in normal human thyroid cells and downregulated in primary thyroid tumors.


Assuntos
Transformação Celular Neoplásica , Regulação para Baixo , Proteínas Ativadoras de GTPase/metabolismo , Proteínas ras/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Ativadoras de GTPase/genética , Humanos , Ratos , Glândula Tireoide/citologia , Glândula Tireoide/metabolismo , Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Proteínas ras/genética
10.
Cancer Res ; 66(21): 10505-12, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17079472

RESUMO

Ras mutations are frequent in thyroid tumors, the most common endocrine malignancy. The ability of Ras to transform thyroid cells is thought to rely on its mitogenic activity. Unexpectedly, acute expression of activated Ras in normal rat thyroid cells induced a DNA damage response, followed by apoptosis. Notably, a subpopulation of cells evaded apoptosis and emerged with features of transformation, including the loss of epithelial morphology, dedifferentiation, and the acquisition of hormone- and anchorage-independent proliferation. Strikingly, the surviving cells showed marked chromosomal instability. Acutely, Ras stimulated replication stress as evidenced by the induction of ataxia telangiectasia mutated and Rad3-related protein kinase (ATR) activity (Chk1 phosphorylation) and of gammaH2A.X, a marker of DNA damage. Despite the activation of a checkpoint, cells continued through mitosis in the face of DNA damage, resulting in an increase in cells harboring micronuclei, an indication of defects in chromosome segregation and other forms of chromosome damage. Cells that survived exposure to Ras continued to exhibit replication stress (ATR activation) but no longer exhibited gammaH2A.X or full activation of p53. When rechallenged with Ras or DNA-damaging agents, the surviving cells were more resistant to apoptosis than parental cells. These data show that acute expression of activated Ras is sufficient to induce chromosomal instability in the absence of other signals, and suggest that Ras-induced chromosomal instability arises as a consequence of defects in the processing of DNA damage. Hence, abrogation of the DNA damage response may constitute a novel mechanism for Ras transformation.


Assuntos
Transformação Celular Neoplásica , Instabilidade Cromossômica , Dano ao DNA , Genes ras/fisiologia , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Reparo do DNA , Histonas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Wistar , Neoplasias da Glândula Tireoide/genética
11.
J Biol Chem ; 281(46): 34759-67, 2006 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-16968694

RESUMO

Genetic evidence indicates that Ras plays a critical role in the initiation and progression of human thyroid tumors. Paradoxically, acute expression of activated Ras in normal rat thyroid cells induced deregulated cell cycle progression and apoptosis. We investigated whether cell cycle progression was required for Ras-stimulated apoptosis. Ras increased CDK-2 activity following its introduction into quiescent cells. Apoptotic cells exhibited a sustained increase in CDK-2 activity, accompanied by the loss of CDK-2-associated p27. Blockade of Ras-induced CDK-2 activity and S phase entry via overexpression of p27 inhibited apoptosis. Inactivation of the retinoblastoma protein in quiescent cells through expression of HPV-E7 stimulated cell cycle progression and apoptosis, indicating that deregulated cell cycle progression is sufficient to induce apoptosis. Ras failed to induce G1 phase growth arrest in normal rat thyroid cells. Rather, Ras-expressing thyroid cells progressed into S and G2 phases and evoked a checkpoint response characterized by the activation of ATR. Ras-stimulated ATR activity, as evidenced by Chk1 and p53 phosphorylation, was blocked by p27, suggesting that cell cycle progression triggers checkpoint activation, likely as a consequence of replication stress. These data reveal that Ras is capable of inducing a DNA damage response with characteristics similar to those reported in precancerous lesions. Our findings also suggest that the frequent mutational activation of Ras in thyroid tumors reflects the ability of Ras-expressing cells to bypass checkpoints and evade apoptosis rather than to simply increase proliferative potential.


Assuntos
Apoptose/fisiologia , Proteínas de Ciclo Celular/metabolismo , Mitógenos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Glândula Tireoide/metabolismo , Proteínas ras/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular , Proteínas de Ciclo Celular/genética , Células Cultivadas , Quinase 1 do Ponto de Checagem , Quinase 2 Dependente de Ciclina/metabolismo , Regulação da Expressão Gênica , Humanos , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Ratos , Ratos Wistar , Glândula Tireoide/citologia , Regulação para Cima , Proteínas ras/genética
12.
Mol Endocrinol ; 20(5): 1112-20, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16410315

RESUMO

We previously reported that protein kinase A activity is an important determinant of thyroid cell survival. Given the important role of cAMP response element binding protein (CREB) in mediating the transcriptional effects of protein kinase A, we explored whether interference with CREB family members impaired thyroid cell survival. Expression of A-CREB, a dominant-negative CREB mutant that inhibits CREB DNA binding activity, induced apoptosis in rat thyroid cells. A-CREB inhibited CRE-regulated gene expression but failed to alter the expression of bcl-2 family members or of well-characterized inhibitors of apoptosis. To elucidate the mechanism through which impaired CREB function triggered apoptosis, its effects on cell proliferation were examined. Expression of A-CREB inhibited cell number increases, in part due to delayed cell cycle transit. Protracted S-phase progression in A-CREB-expressing cells was sufficient to activate a checkpoint response characterized by Chk-1, histone H2A.X, and p53 phosphorylation. To determine whether cell cycle progression was required for apoptosis, the effects of p27 overexpression were investigated. Overexpression of p27 prevented cell cycle progression, checkpoint activation, and apoptosis in A-CREB-expressing cells. These data reveal a novel mechanism through which interference with CREB abrogates cell survival, through checkpoint activation secondary to cell cycle delay. This study may explain how interference with CREB induces apoptosis in cells where alterations in the expression of pro- and anti-survival genes are not detected.


Assuntos
Apoptose , Ciclo Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/antagonistas & inibidores , Proteínas Quinases/metabolismo , Animais , Apoptose/genética , Ciclo Celular/genética , Células Cultivadas , Quinase 1 do Ponto de Checagem , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Expressão Gênica , Histonas/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Glândula Tireoide/citologia , Glândula Tireoide/metabolismo , Proteína Supressora de Tumor p53/metabolismo
13.
J Biol Chem ; 280(37): 32107-14, 2005 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-16051606

RESUMO

Overexpression of protein kinase C delta (PKCdelta) stimulates apoptosis in a wide variety of cell types through a mechanism that is incompletely understood. PKCdelta-deficient cells are impaired in their response to DNA damage-induced apoptosis, suggesting that PKCdelta is required to mount an appropriate apoptotic response under conditions of stress. The mechanism through which it does so remains elusive. In addition to effects on cell survival, PKCdelta elicits pleiotropic effects on cellular proliferation. We now provide the first evidence that the ability of PKCdelta to stimulate apoptosis is intimately linked to its ability to stimulate G(1) phase cell cycle progression. Using an adenoviral-based expression system to express PKCalpha,-delta, and -epsilon in epithelial cells, we demonstrate that a modest increase in PKCdelta activity selectively stimulates quiescent cells to initiate G(1) phase cell cycle progression. Rather than completing the cell cycle, PKCdelta-infected cells arrest in S phase, an event that triggers caspase-dependent apoptotic cell death. Apoptosis was preceded by the activation of cell cycle checkpoints, culminating in the phosphorylation of Chk-1 and p53. Strikingly, blockade of S phase entry using the phosphatidylinositol 3-kinase inhibitor LY294002 prevented checkpoint activation and apoptosis. In contrast, inhibitors of mitogen-activated protein kinase cascades failed to prevent apoptosis. These findings demonstrate that the biological effects of PKCdelta can be extended to include positive regulation of G(1) phase cell cycle progression. Importantly, they reveal the existence of a novel, cell cycle-dependent mechanism through which PKCdelta stimulates cell death.


Assuntos
Apoptose , Proteína Quinase C/fisiologia , Adenoviridae/genética , Animais , Western Blotting , Bromodesoxiuridina/farmacologia , Ciclo Celular , Morte Celular , Proliferação de Células , Quinase 1 do Ponto de Checagem , Cromonas/farmacologia , DNA/metabolismo , Dano ao DNA , Inibidores Enzimáticos/farmacologia , Células Epiteliais/metabolismo , Citometria de Fluxo , Fase G1 , Modelos Biológicos , Morfolinas/farmacologia , Fosforilação , Proteína Quinase C/metabolismo , Proteína Quinase C-delta , Proteínas Quinases/metabolismo , Ratos , Ratos Wistar , Fase S , Glândula Tireoide/citologia , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
14.
Thyroid ; 15(2): 114-20, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15753668

RESUMO

Feline hyperthyroidism is a common, spontaneous disease in older cats that is similar clinically and histopathologically to human toxic multinodular goiter (TNG). In this study, the functional response of feline normal thyroid (NT) and hyperthyroid (HT) cells grown in monolayer culture to thyrotropin (TSH) was determined. Basal levels of DNA synthesis were similar in NT and HT cells. TSH stimulated concentration-dependent DNA synthesis in NT and HT cells, with maximal stimulation seen at 1 and 10 mU/mL TSH in NT and HT cells, respectively. HT cells had higher basal levels of thyroglobulin (Tg) expression. TSH stimulated Tg expression in NT and HT cells in a concentration-dependent fashion, with maximal activity at 0.5 and 5 mU/mL TSH, respectively. These results demonstrate that NT and HT cells in monolayer culture exhibit growth and functional responses to TSH. HT cells have higher basal Tg expression than NT cells and require higher TSH concentrations to stimulate DNA synthesis and Tg expression, two measures of thyroid cell activation. These data support the idea that feline hyperthyroidism is caused by cell abnormalities, resulting in dysregulated growth and hormone synthesis, and emphasize its importance as an animal model for TNG.


Assuntos
Doenças do Gato/fisiopatologia , Bócio Nodular/veterinária , Hipertireoidismo/veterinária , Tireoglobulina/metabolismo , Tireotropina/farmacologia , Animais , Western Blotting , Doenças do Gato/metabolismo , Gatos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura/farmacologia , DNA/biossíntese , Bócio Nodular/metabolismo , Bócio Nodular/fisiopatologia , Hipertireoidismo/metabolismo , Hipertireoidismo/fisiopatologia , Glândula Tireoide/citologia , Glândula Tireoide/metabolismo
15.
Mol Endocrinol ; 18(9): 2321-32, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15166254

RESUMO

Thyroid cell proliferation is regulated by the concerted action of TSH/cAMP and serum growth factors. The specific contributions of cAMP-dependent vs. -independent signals to cell cycle progression are not well understood. We examined the molecular basis for the synergistic effects of TSH and serum on G1/S phase cell cycle progression in rat thyroid cells. Although strictly required for thyroid cell proliferation, TSH failed to stimulate G1 phase cell cycle progression. Together with serum, TSH increased the number of cycling cells. TSH enhanced the effects of serum on retinoblastoma protein hyperphosphorylation, cyclin-dependent kinase 2 activity, and cyclin A expression. Most notably, TSH and serum elicited strikingly different effects on p27 localization. TSH stimulated the nuclear accumulation of p27, whereas serum induced its nuclear export. Unexpectedly, TSH enhanced the depletion of nuclear p27 in serum-treated cells. Furthermore, only combined treatment with TSH and serum led to rapamycin-sensitive p27 turnover. Together, TSH and serum stimulated p70S6K activity that remained high through S phase. These data suggest that TSH regulates cell cycle progression, in part, by increasing the number of cycling cells through p70S6K-mediated effects on the localization of p27.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Soro/fisiologia , Glândula Tireoide/metabolismo , Tireotropina/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Quinases relacionadas a CDC2 e CDC28/metabolismo , Bovinos , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/análise , Núcleo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Colforsina/farmacologia , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27 , DNA/análise , DNA/biossíntese , Fosforilação , Transporte Proteico/fisiologia , Ratos , Proteína do Retinoblastoma/metabolismo , Glândula Tireoide/citologia , Tireotropina/fisiologia , Proteínas Supressoras de Tumor/análise
16.
Cancer Res ; 64(3): 1079-86, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-14871841

RESUMO

Valproic acid (VPA), a well-established therapy for seizures and bipolar disorder, has recently been shown to inhibit histone deacetylases (HDACs). Similar to more widely studied HDAC inhibitors, VPA can cause growth arrest and induce differentiation of transformed cells in culture. Whether this effect of VPA is through inhibition of HDACs or modulation of another target of VPA has not been tested. We have used a series of VPA analogs to establish a pharmacological profile for HDAC inhibition. We find that VPA and its analogs inhibit multiple HDACs from class I and class II (but not HDAC6 or HDAC10) with a characteristic order of potency in vitro. These analogs also induce hyperacetylation of core histones H3 and H4 in intact cells with an order of potency that parallels in vitro inhibition. VPA and VPA analogs induce differentiation in hematopoietic cell lines in a p21-dependent manner, and the order of potency for induction of differentiation parallels the potencies for inhibition in vitro, as well as for acetylation of histones associated with the p21 promoter, supporting the argument that differentiation caused by VPA is mediated through inhibition of HDACs. These findings provide additional evidence that VPA, a well-tolerated, orally administered drug with extensive clinical experience, may serve as an effective chemotherapeutic agent through targeting of HDACs.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Ácido Valproico/análogos & derivados , Ácido Valproico/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/fisiologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Isoenzimas/antagonistas & inibidores , Células K562 , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células U937
17.
J Biol Chem ; 279(7): 5501-7, 2004 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-14660640

RESUMO

Beyond regulating Rap activity, little is known regarding the regulation and function of the Rap GTPase-activating protein Rap1GAP. Tuberin and E6TP1 protein levels are tightly regulated through ubiquitin-mediated proteolysis. A role for these RapGAPs, along with SPA-1, as tumor suppressors has been demonstrated. Whether Rap1GAP performs a similar role was investigated. We now report that Rap1GAP protein levels are dynamically regulated in thyroid-stimulating hormone (TSH)-dependent thyroid cells. Upon TSH withdrawal, Rap1GAP undergoes a net increase in phosphorylation followed by proteasome-mediated degradation. Sequence analysis identified two putative destruction boxes in the Rap1GAP C-terminal domain. Glycogen synthase kinase 3beta (GSK3beta) phosphorylated Rap1GAP immunoprecipitated from thyroid cells, and GSK3beta inhibitors prevented phosphorylation and degradation of endogenous Rap1GAP. Co-expression of GSK3beta and Rap1GAP in human embryonic kidney 293 cells stimulated proteasome-dependent Rap1GAP turnover. Mutational analysis established a role for serine 525 in the regulation of Rap1GAP stability. Overexpression of Rap1GAP in thyroid cells impaired TSH/cAMP-stimulated p70S6 kinase activity and cell proliferation. These data are the first to show that Rap1GAP protein levels are tightly regulated and are the first to support a role for Rap1GAP as a tumor suppressor.


Assuntos
AMP Cíclico/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Tireotropina/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Western Blotting , Bromodesoxiuridina/farmacologia , Divisão Celular , Linhagem Celular , Cisteína Endopeptidases/metabolismo , Análise Mutacional de DNA , Relação Dose-Resposta a Droga , Proteínas Ativadoras de GTPase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Complexos Multienzimáticos/antagonistas & inibidores , Complexos Multienzimáticos/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Fosforilação , Testes de Precipitina , Complexo de Endopeptidases do Proteassoma , Estrutura Terciária de Proteína , Ratos , Ratos Wistar , Proteínas Repressoras/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Glândula Tireoide/metabolismo , Fatores de Tempo , Transfecção , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor
19.
J Neuropathol Exp Neurol ; 62(9): 936-50, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14533783

RESUMO

The mechanisms whereby mutant gene expression triggers neurodegeneration are poorly understood but have generally been attributed to translated gene products. We now demonstrate direct neuropathic effects of untranslated RNA on cultured motor neurons. We show that expression of untranslated light neurofilament (NF-L) RNA sequence in the 3'UTR of an EGFP transgene (pEGFP/NF-L RNA) or in a separate expression vector (pRc/NF-L RNA) causes dose-dependent, neuron-specific motor neuron degeneration. Neither unfused EGFP protein (pEGFP/wt) nor EGFP-tagged NF-L protein (pEGFP/NF-L protein) has similar neuropathic effects. The findings are the first demonstration of a direct RNA-mediated neurotoxic effect. Moreover, the resulting neuropathological changes show that untranslated RNA can lead to early degeneration of neuritic processes and accumulations of ubiquitinated aggregates in the perikarya and nuclei of degenerating motor neurons. The latter findings are hallmark neuropathological features of neurodegenerative diseases and their occurrence as a result of altered RNA expression raises the prospects of an RNA-mediated component in the pathogenesis of neurodegenerative states.


Assuntos
Neurônios Motores/metabolismo , Degeneração Neural/metabolismo , Proteínas de Neurofilamentos/biossíntese , RNA/biossíntese , Ubiquitinas/biossíntese , Animais , Agregação Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Camundongos , Neurônios Motores/patologia , Degeneração Neural/patologia , Proteínas de Neurofilamentos/genética , RNA/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia , Ubiquitinas/genética
20.
Mol Endocrinol ; 17(3): 450-9, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12554771

RESUMO

Abundant evidence supports the ability of Ras to stimulate thyroid cell proliferation. Stable expression of activated Ras enhances the sensitivity of thyroid cells to apoptosis. We report that apoptosis is a primary and general response of rat thyroid cells to acute expression of activated Ras in the absence or presence of thyrotropin, insulin, and serum, survival factors for thyroid cells. Ras induced apoptosis in quiescent and cycling cells. Concomitantly, Ras stimulated S phase entry in quiescent cells and enhanced G1/S transition in cycling cells. Ras effects on the cell cycle were characterized by delayed progression through S phase and an apparent failure to proceed through G2/M phase. Unlike thyroid cell mitogens, Ras markedly decreased cyclin D1 expression. Although acute expression of Ras decreased cyclin D1 protein levels, cells selected to survive chronic Ras expression exhibited a selective increase in cyclin D1 expression. In summary, thyroid cells harbor an apoptotic program activated by Ras that outstrips the protective effects of thyrotropin, insulin, and serum. Apoptosis is accompanied by dysregulated cell cycle progression, suggesting that cell death may arise, at least in part, as a consequence of inappropriate proliferative cues.


Assuntos
Apoptose/fisiologia , Ciclo Celular/fisiologia , Glândula Tireoide/citologia , Proteínas ras/fisiologia , Animais , Western Blotting , Bromodesoxiuridina/metabolismo , Células Cultivadas , Ciclina D1/genética , Ciclina D1/fisiologia , Citometria de Fluxo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA