Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Autophagy ; 10(6): 1071-92, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24879154

RESUMO

DIRAS3 is an imprinted tumor suppressor gene that is downregulated in 60% of human ovarian cancers. Re-expression of DIRAS3 at physiological levels inhibits proliferation, decreases motility, induces autophagy, and regulates tumor dormancy. Functional inhibition of autophagy with choroquine in dormant xenografts that express DIRAS3 significantly delays tumor regrowth after DIRAS3 levels are reduced, suggesting that autophagy sustains dormant ovarian cancer cells. This study documents a newly discovered role for DIRAS3 in forming the autophagosome initiation complex (AIC) that contains BECN1, PIK3C3, PIK3R4, ATG14, and DIRAS3. Participation of BECN1 in the AIC is inhibited by binding of BECN1 homodimers to BCL2. DIRAS3 binds BECN1, disrupting BECN1 homodimers and displacing BCL2. Binding of DIRAS3 to BECN1 increases the association of BECN1 with PIK3C3 and ATG14, facilitating AIC activation. Amino acid starvation of cells induces DIRAS3 expression, reduces BECN1-BCL2 interaction and promotes autophagy, whereas DIRAS3 depletion blocks amino acid starvation-induced autophagy. In primary ovarian cancers, punctate expression of DIRAS3, BECN1, and the autophagic biomarker MAP1LC3 are highly correlated (P<0.0001), underlining the clinical relevance of these mechanistic studies. Punctate expression of DIRAS3 and MAP1LC3 was detected in only 21-23% of primary ovarian cancers but in 81-84% of tumor nodules found on the peritoneal surface at second-look operations following primary chemotherapy. This reflects a 4-fold increase (P<0.0001) in autophagy between primary disease and post-treatment recurrence. We suggest that DIRAS3 not only regulates the AIC, but induces autophagy in dormant, nutrient-deprived ovarian cancer cells that remain after conventional chemotherapy, facilitating their survival.


Assuntos
Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Aminoácidos/metabolismo , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/genética , Autofagia/fisiologia , Proteína 12 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Proteína Beclina-1 , Linhagem Celular Tumoral , Feminino , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Pessoa de Meia-Idade , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neoplasias Ovarianas/genética , Fagossomos/metabolismo , Fagossomos/patologia , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Proteínas rho de Ligação ao GTP/química , Proteínas rho de Ligação ao GTP/genética
2.
Int J Pept Res Ther ; 19(1): 3-12, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24707243

RESUMO

Signal transducer and activator of transcription 3 (Stat3) transmits signals from growth factors and interleukin-6 family cytokines by binding to their receptors via its Src homology 2 (SH2) domain. This results in phosphorylation of Tyr705, dimerization, translocation to the nucleus, and regulation of transcription of downstream genes. Stat3 is constitutively activated in several human cancers and is a target for anti-cancer drug design. We have shown previously phosphorylation of Tyr705 in intact cancer cells can be inhibited with prodrugs of phosphopeptide mimics targeting the SH2 domain. In a series of prodrugs consisting of bis-pivaloyloxymethyl esters of 4'-phosphonodifluoromethyl cinnamoyl-Haic-Gln-NHBn, appending methyl group to the ß-position of the cinnamate increased potency ca. twofold, which paralleled the increase in affinity of the corresponding phosphopeptide models. However, dramatic increases in potency were observed when the C-terminal C(O)NHBn of Gln-NHBn was replaced with a simple methyl group. In this communication we continue to explore the effects of structural modifications of prodrugs on their ability to inhibit Tyr705 phosphorylation. A set of 4-substituted prolines incorporated into ß-methyl-4-phosphocinnamoyl-leucinyl-Xaa-4-aminopentamide model peptides exhibited affinities of 88-317 nM by fluorescence polarization (Pro IC50 = 156 nM). In corresponding prodrugs, Pro inhibited constitutive Stat3 phosphorylation at 10 µM in MDA-MB-468 breast tumor cells. However, 4,4-difluoroproline and 4,4-dimethylproline resulted in complete inhibition at 0.5 µM. These results suggest that the prodrug with native proline undergoes metabolism that those with substituted prolines do not. In conclusion, changes in structure with minimal impact on intrinsic affinity can nevertheless have profound effects on the cellular potency of prodrug inhibitors of Stat3.

3.
J Exp Ther Oncol ; 10(2): 155-62, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23350355

RESUMO

Signal transducer and activator of transcription 3 (Stat3) is constitutively activated in a number of human cancers and cancer cell lines. Via its Src homology 2 (SH2) domain, Stat3 is recruited to phosphotyrosine residues on intracellular domains of cytokine and growth factor receptors, whereupon it is phosphorylated on Tyr705, dimerizes, translocates to the nucleus and is reported to participate in the expression of genes related to angiogenesis, metastasis, growth and survival. To block this process, we are developing cell-permeable, phosphatase-stable phosphopeptide mimics, targeted to the SH2 domain of Stat3, that inhibit the phosphorylation of Tyr705 of Stat3 in cultured tumor cells (Mandal et al., J. Med. Chem. 54, 3549-5463, 2011). At concentrations that inhibit tyrosine phosphorylation, these materials were not cytotoxic, similar to recent reports on JAK inhibitors. At higher concentrations, cytotoxicity was accompanied by off-target effects. We report that treatment of MDA-MB-468 human breast cancer xenografts in mice with peptidomimetic PM-73G significantly inhibited tumor growth, which was accompanied by reduction in VEGF production and microvessel density. No evidence of apoptosis or changes in the expression of the canonical genes cyclin D1 or survivin were observed. Thus selective inhibition of Stat3 Tyr705 phosphorylation may be a novel anti-angiogenesis strategy for the treatment of cancer.


Assuntos
Biomimética , Neoplasias da Mama/prevenção & controle , Neovascularização Patológica/prevenção & controle , Fosfopeptídeos/farmacologia , Pró-Fármacos/farmacologia , Fator de Transcrição STAT3/metabolismo , Domínios de Homologia de src/efeitos dos fármacos , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Feminino , Humanos , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
JAKSTAT ; 1(4): 263-347, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24058783

RESUMO

Herein we review our progress on the development of phosphopeptide-based prodrugs targeting the SH2 domain of STAT3 to prevent recruitment to cytokine and growth factor receptors, activation, nuclear translocation and transcription of genes involved in cancer. We developed high affinity phosphopeptides (K I = 46-200 nM). Corresponding prodrugs inhibited constitutive and IL-6 induced Tyr705 phosphorylation at 0.5-1 µM in a variety of human cancer cell lines. They were not cytotoxic at 5 µM in vitro but they inhibited tumor growth in a human xenograft breast cancer model in mice, accompanied by reduced VEGF expression and angiogenesis.

5.
Breast Cancer Res ; 13(3): R65, 2011 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-21689417

RESUMO

INTRODUCTION: The c-Jun coactivator, Jun activation-domain binding protein 1 (Jab1) also known as the fifth component of the COP9 signalosome complex (CSN5), is a novel candidate oncogene whose aberrant expression contributes to the progression of breast carcinoma and other human cancers. The mechanism of Jab1 gene expression and its deregulation in cancer cells remains to be identified. We therefore investigated the transcriptional regulatory mechanisms of Jab1 expression in human breast carcinoma cells. METHODS: To identify potential regulators of Jab1 transcription, we cloned the 5' upstream region of the human Jab1 gene and mapped its transcriptional start site. We identified binding sequences for the CCAAT/enhancer binding protein (C/EBP) and GATA, as well as a signal transducer and activator of transcription-3 (Stat3) consensus sequence overlapping the C/EBP site, using 5'- deletion analysis and a gene reporter assay. Mutational analysis of these binding sites was performed to confirm their roles in promoting Jab1 transcription in breast cancer cells. We further confirmed these binding sites using electrophoretic mobility shift assays (EMSAs) and chromatin immunoprecipitation (ChIP) assays. We also analyzed whether the siRNA-mediated inactivation of Stat3 and Src could reduce Jab1-promoter activity and whether interleukine-6 (IL-6) could mediate increased Jab1 expression through Stat3 signaling. RESULTS: We identified binding sequences for C/EBP, GATA, as well as a Stat3 consensus sequence overlapping the C/EBP site in the promoter region of Jab1. C/EBP-beta2 is a potential transcriptional activator of Jab1 and mutation of the C/EBP/Stat3 binding site significantly reduced Jab1-promoter activity. In addition, inhibiting Stat3 significantly reduced Jab1-promoter activation. EMSA and ChIP assays confirmed that C/EBP, GATA1 and Stat3 bind to Jab1 promoter in breast carcinoma cells. We also found that Src, an activator of Stat3, is involved in Jab1-promoter activation. siRNA knockdown of Src reduced the Jab1-promoter activity, similar to the results seen when Stat3 was inhibited in breast carcinoma cells. Interestingly, reactivation of Stat3 in normal mammary epithelial cells (MCF-10A, MCF-10F) is sufficient to reactivate Jab1 expression. Treatment with the cytokine IL-6 resulted in increased Jab1 expression that was blocked by inhibition of Stat3. CONCLUSIONS: These findings reveal a novel mechanism of Jab1 gene regulation and provide functional and mechanistic links between the Src/Stat3 and IL-6/Stat3 signaling axes that are involved in the activation of Jab1 transcription and regulation of this novel oncogenic protein.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeo Hidrolases/genética , Regiões Promotoras Genéticas , Fator de Transcrição STAT3/metabolismo , Sequência de Bases , Sítios de Ligação , Proteína beta Intensificadora de Ligação a CCAAT , Complexo do Signalossomo COP9 , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Fator de Transcrição GATA1/metabolismo , Humanos , Interleucina-6/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeo Hidrolases/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Sequências Reguladoras de Ácido Nucleico , Fator de Transcrição STAT3/antagonistas & inibidores , Análise de Sequência de DNA , Transdução de Sinais/genética , Transcrição Gênica , Ativação Transcricional , Quinases da Família src/genética , Quinases da Família src/metabolismo
6.
Cancer ; 117(19): 4424-38, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21491416

RESUMO

BACKGROUND: Epigenetic therapy has had a significant impact on the management of hematologic malignancies, but its role in the treatment of ovarian cancer remains to be defined. The authors previously demonstrated that treatment of ovarian and breast cancer cells with DNA methyltransferase and histone deacetylase (HDAC) inhibitors can up-regulate the expression of imprinted tumor suppressors. In this study, demethylating agents and HDAC inhibitors were tested for their ability to induce re-expression of tumor suppressor genes, inhibiting growth of ovarian cancer cells in culture and in xenografts. METHODS: Ovarian cancer cells (Hey and SKOv3) were treated with demethylating agents (5-aza-20-deoxycytidine [DAC] or 5-azacitidine [AZA]) or with HDAC inhibitors (suberoylanilide hydroxamicacid [SAHA] or trichostatin A [TSA]) to determine their impact on cellular proliferation, cell cycle regulation, apoptosis, autophagy, and re-expression of 2 growth inhibitory imprinted tumor suppressor genes: guanosine triphosphate-binding Di-RAS-like 3 (ARHI) and paternally expressed 3 (PEG3). The in vivo activities of DAC and SAHA were assessed in a Hey xenograft model. RESULTS: The combination of DAC and SAHA produced synergistic inhibition of Hey and SKOv3 cell growth by apoptosis and cell cycle arrest. DAC induced autophagy in Hey cells that was enhanced by SAHA. Treatment with both agents induced re-expression of ARHI and PEG3 in cultured cells and in xenografts, correlating with growth inhibition. Knockdown of ARHI decreased DAC-induced autophagy. DAC and SAHA inhibited the growth of Hey xenografts and induced autophagy in vivo. CONCLUSIONS: A combination of DAC and SAHA inhibited ovarian cancer growth while inducing apoptosis, G2/M arrest, autophagy, and re-expression of imprinted tumor suppressor genes.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia , Azacitidina/análogos & derivados , Genes Supressores de Tumor/efeitos dos fármacos , Impressão Genômica , Ácidos Hidroxâmicos/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Animais , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Decitabina , Sinergismo Farmacológico , Quimioterapia Combinada , Epigenômica , Feminino , Fase G2/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Transplante Heterólogo , Vorinostat
7.
J Med Chem ; 54(10): 3549-63, 2011 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-21486047

RESUMO

Signal transducer and activator of transcription 3 (Stat3), a target for anticancer drug design, is activated by recruitment to phosphotyrosine residues on growth factor and cytokine receptors via its SH2 domain. We report here structure-activity relationship studies on phosphopeptide mimics targeted to the SH2 domain of Stat3. Inclusion of a methyl group on the ß-position of the pTyr mimic 4-phosphocinnamide enhanced affinity 2- to 3-fold. Bis-pivaloyloxymethyl prodrugs containing ß-methylcinnamide, dipeptide scaffolds Haic and Nle-cis-3,4-methanoproline, and glutamine surrogates were highly potent, completely inhibiting phosphorylation of Stat3 Tyr705 at 0.5-1 µM in a variety of cancer cell lines. The inhibitors were selective for Stat3 over Stat1, Stat5, Src, and p85 of PI3K, indicating ability to discriminate individual SH2 domains in intact cells. At concentrations that completely inhibited Stat3 phosphorylation, the prodrugs were not cytotoxic to a panel of tumor cells, thereby showing clear distinction between cytotoxicity and effects downstream of activated Stat3.


Assuntos
Química Farmacêutica/métodos , Fator de Transcrição STAT3/química , Domínios de Homologia de src , Transporte Ativo do Núcleo Celular , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Desenho de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Cinética , Modelos Químicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Pró-Fármacos , Estrutura Terciária de Proteína , Transdução de Sinais
8.
Org Lett ; 11(15): 3394-7, 2009 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-19594124

RESUMO

The synthesis of prodrugs targeted to the SH2 domain of Stat3 is reported. Using a convergent strategy, the pivaloyloxymethyl phosphonodiester of pentachlorophenyl 4-phosphonodifluoromethylcinnamate, a phosphotyrosine surrogate, was synthesized and used to acylate peptidomimetic fragments that were prepared on solid supports. Two prodrugs described here inhibited the phosphorylation of Stat3 in breast tumor cells.


Assuntos
Antineoplásicos/síntese química , Compostos Heterocíclicos com 3 Anéis/síntese química , Pró-Fármacos/síntese química , Pirrolidinas/síntese química , Fator de Transcrição STAT3/antagonistas & inibidores , Domínios de Homologia de src/efeitos dos fármacos , Antineoplásicos/química , Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Dimerização , Feminino , Compostos Heterocíclicos com 3 Anéis/química , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Fosforilação/efeitos dos fármacos , Fosfotirosina/síntese química , Fosfotirosina/química , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Pirrolidinas/química , Pirrolidinas/farmacologia
9.
Cancer ; 115(17): 3897-908, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19517469

RESUMO

BACKGROUND: Constitutively activated nuclear factor kappa B (NFkappaB) contributes to the development of cancer by regulating the expression of genes involved in cell survival, metastasis, and angiogenesis. The authors have demonstrated that MEKK3 plays a critical role in cytokine-mediated NFkappaB activation, and that stable expression of MEKK3 in cultured cells leads to increased NFkappaB activity. METHODS: MEKK3 expression in ovarian cancer cells or tumors was assessed by Western blotting and real-time polymerase chain reaction. NFkappaB activities were analyzed by electrophoretic mobility shift assay and luciferase reporter assays. Western blot analysis for the survival factors were also performed and correlated with MEKK3 and NFkappaB activities. Cell survival assays were used to determine the sensitivity of ovarian cancer cells to various chemotherapeutic agents. RESULTS: The authors found that 63% of the ovarian cancers had higher MEKK3 expression than the normal ovarian epithelial cells. Ovarian cancers with high MEKK3 showed correspondingly high IkappaB kinase and NFkappaB activity. Moreover, MEKK3 coimmunoprecipitated with Akt and cooperated with Akt to synergistically activate NFkappaB. Consistent with increased MEKK3 and NFkappaB activity in ovarian cancers, Bcl-2, Bcl-xL, survivin, and X-linked inhibitor of apoptosis levels were increased, which correlated with increased resistance to chemotherapeutic agents. Knockdown of MEKK3 with small interfering RNA significantly increased cancer cell sensitivity to paclitaxel. CONCLUSIONS: MEKK3 may be aberrantly expressed in ovarian cancers and plays an important role in tumors with constitutively activated NFkappaB.


Assuntos
Apoptose/genética , Cistadenocarcinoma Seroso/enzimologia , Cistadenocarcinoma Seroso/genética , MAP Quinase Quinase Quinase 3/metabolismo , NF-kappa B/metabolismo , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/genética , Linhagem Celular Tumoral , Feminino , Humanos , Interleucina-1 , Proteínas Proto-Oncogênicas c-akt/metabolismo
10.
Biosci Rep ; 30(3): 159-68, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19435463

RESUMO

ARHI (aplasia Ras homologue member I; also known as DIRAS3) is an imprinted tumour suppressor gene, the expression of which is lost in the majority of breast and ovarian cancers. Unlike its homologues Ras and Rap, ARHI functions as a tumour suppressor. Our previous study showed that ARHI can interact with the transcriptional activator STAT3 (signal transducer and activator of transcription 3) and inhibit its nuclear translocation in human breast- and ovarian-cancer cells. To identify proteins that interact with ARHI in nuclear translocation, in the present study, we performed proteomic analysis and identified several importins that can associate with ARHI. To further explore this novel finding, we purified 10 GST (glutathione transferase)-importin fusion proteins (importins 7, 8, 13, beta1, alpha1, alpha3, alpha5, alpha6, alpha7 and mutant alpha1). Using a GST-pulldown assay, we found that ARHI can bind strongly to most importins; however, its binding is markedly reduced with an importin alpha1 mutant that contains an altered NLS (nuclear-localization signal) domain. In addition, an ARHI N-terminal deletion mutant exhibits greatly reduced binding to all importins compared with wild-type ARHI. In nuclear-import assays, the addition of ARHI blocked nuclear localization of phosphorylated STAT3. ARHI also inhibits the interaction of Ran-importin complexes with GFP (green fluorescent protein) fusion proteins that contain an NLS domain and a beta-like import receptor-binding domain, thereby blocking their nuclear localization. By conducting GST-pulldown assays, we found that ARHI could compete for Ran-importin binding. Thus ARHI-induced disruption of importin-binding to cargo proteins, including STAT3, could serve as an important regulatory mechanism that contributes to the tumour-suppressor function of ARHI.


Assuntos
Núcleo Celular/metabolismo , Carioferinas/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo , Transporte Ativo do Núcleo Celular , Western Blotting , Linhagem Celular Tumoral , Genes Supressores de Tumor , Impressão Genômica , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Carioferinas/genética , Microscopia de Fluorescência , Sinais de Localização Nuclear/genética , Ligação Proteica , Transporte Proteico , Proteômica/métodos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição STAT3/genética , Proteína ran de Ligação ao GTP/genética , Proteína ran de Ligação ao GTP/metabolismo
12.
J Clin Invest ; 118(12): 3917-29, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19033662

RESUMO

The role of autophagy in oncogenesis remains ambiguous, and mechanisms that induce autophagy and regulate its outcome in human cancers are poorly understood. The maternally imprinted Ras-related tumor suppressor gene aplasia Ras homolog member I (ARHI; also known as DIRAS3) is downregulated in more than 60% of ovarian cancers, and here we show that re-expression of ARHI in multiple human ovarian cancer cell lines induces autophagy by blocking PI3K signaling and inhibiting mammalian target of rapamycin (mTOR), upregulating ATG4, and colocalizing with cleaved microtubule-associated protein light chain 3 (LC3) in autophagosomes. Furthermore, ARHI is required for spontaneous and rapamycin-induced autophagy in normal and malignant cells. Although ARHI re-expression led to autophagic cell death when SKOv3 ovarian cancer cells were grown in culture, it enabled the cells to remain dormant when they were grown in mice as xenografts. When ARHI levels were reduced in dormant cells, xenografts grew rapidly. However, inhibition of ARHI-induced autophagy with chloroquine dramatically reduced regrowth of xenografted tumors upon reduction of ARHI levels, suggesting that autophagy contributed to the survival of dormant cells. Further analysis revealed that autophagic cell death was reduced when cultured human ovarian cancer cells in which ARHI had been re-expressed were treated with growth factors (IGF-1, M-CSF), angiogenic factors (VEGF, IL-8), and matrix proteins found in xenografts. Thus, ARHI can induce autophagic cell death, but can also promote tumor dormancy in the presence of factors that promote survival in the cancer microenvironment.


Assuntos
Autofagia , Neoplasias Ovarianas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Antirreumáticos/farmacologia , Autofagia/efeitos dos fármacos , Autofagia/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cloroquina/farmacologia , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Feminino , Impressão Genômica/efeitos dos fármacos , Impressão Genômica/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Transplante de Neoplasias , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Fagossomos/genética , Fagossomos/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Transplante Heterólogo , Proteínas Supressoras de Tumor/genética , Proteínas rho de Ligação ao GTP/genética
13.
Circ Res ; 102(8): 933-41, 2008 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-18309099

RESUMO

Homocysteine (Hcy) contributes to atherogenesis and angiostasis by altering the phenotype of arterial endothelial cells (ECs). The present study was aimed at elucidating potential mechanisms by which Hcy can slow EC proliferation and induce EC apoptosis, thereby disrupting endothelial integrity. Given the strong mitogenic and antiapoptotic properties of fibroblast growth factor (FGF)2, we examined whether Hcy can modulate its expression. In cultured human coronary and bovine aortic ECs, Hcy exerted time- and concentration-dependent (0 to 500 micromol/L) reduction of the mRNA and protein levels of FGF2, whereas vascular endothelial growth factor expression was not affected until Hcy reached a proapoptotic 500 micromol/L. By testing a panel of signal transduction inhibitors, we found that the Hcy-induced downregulation of FGF2 was specifically attenuated by pertussis toxin, an inhibitor of Gi protein signaling. Hcy induced cell cycle arrest at the G(1)/S transition and increased TUNEL-positive apoptotic cells in a graded manner. These effects were effectively counteracted by exogenous FGF2. Reporter gene assays showed that Hcy downregulated FGF2 by transcriptional repression of the gene promoter encompassed in a CpG dinucleotide-rich island. This region was heavily methylated at the cytosine residues by Hcy despite decreased methylation potential (S-adenosylmethionine to S-adenosylhomocysteine ratio). Normal levels of FGF2 transcription were restored to ECs simultaneously exposed to Hcy and 5-aza-deoxycytidine. We conclude that homocysteine disrupts the growth and survival of ECs through a G protein-mediated pathway associated with altered promoter DNA methylation and the transcriptional repression of FGF2.


Assuntos
Artérias/citologia , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/citologia , Fator 2 de Crescimento de Fibroblastos/genética , Proteínas de Ligação ao GTP/metabolismo , Homocistina/farmacologia , Animais , Bovinos , Metilação de DNA , Regulação para Baixo/efeitos dos fármacos , Transcrição Gênica
14.
Diabetes ; 57(1): 158-66, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17959932

RESUMO

OBJECTIVE: L5, a circulating electronegative LDL identified in patients with hypercholesterolemia or type 2 diabetes, induces endothelial cell (EC) apoptosis by suppressing fibroblast growth factor (FGF)2 expression. FGF2 plays a pivotal role in endothelial regeneration and compensatory arteriogenesis. It is likely that vasculopathy and poor collateralization in diabetes is a result of FGF2 dysregulation. RESEARCH DESIGN AND METHODS: To investigate this mechanism, we isolated L5 from type 2 diabetic patients. In cultured bovine aortic ECs (BAECs), L5 inhibited FGF2 transcription and induced apoptosis. Because FGF2 stimulates the phosphatidylinositol 3-kinase (PI3K)-Akt pathway, we examined whether FGF2 transcription is regulated by Akt through a feedback mechanism. RESULTS: Diabetic L5 reduced FGF2 release to the medium but enhanced caspase-3 activity, with resultant apoptosis. Inhibition of PI3K with wortmannin or suppression of Akt activation with dominant-negative Akt inhibited FGF2 expression. Transfection of BAECs with FGF2 antisense cDNA depleted endogenous FGF2 protein. In these cells, not only was Akt phosphorylation inhibited, but FGF2 transcription was also critically impaired. In contrast, transfecting BAECs with FGF2 sense cDNA augmented Akt phosphorylation. Treatment with constitutively active Akt enhanced FGF2 expression. Augmentation of either FGF2 transcription or Akt phosphorylation rendered BAECs resistant to L5. CONCLUSIONS: These findings suggest that FGF2 is the primary initiator of its own expression, which is autoregulated through a novel FGF2-PI3K-Akt loop. Thus, by disrupting FGF2 autoregulation in vascular ECs, L5 may impair reendothelialization and collateralization in diabetes.


Assuntos
Apoptose/efeitos dos fármacos , Diabetes Mellitus Tipo 2/fisiopatologia , Endotélio Vascular/fisiologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Lipoproteínas LDL/sangue , Animais , Aorta , Bovinos , Diabetes Mellitus Tipo 2/patologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Homeostase , Humanos , Lipoproteínas LDL/isolamento & purificação , Lipoproteínas LDL/farmacologia , Valores de Referência , Fator A de Crescimento do Endotélio Vascular/farmacologia
15.
Int J Cancer ; 120(8): 1664-8, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17230502

RESUMO

ARHI is a maternally imprinted tumor suppressor gene that is expressed in normal breast and ovarian epithelial cells but not in most breast and ovarian cancers. Our earlier studies showed that histone deacetylases (HDACs) in complexes with transcription factors E2F1 and E2F4 play an important role in downregulating ARHI expression in breast cancer cells. To determine which HDAC or HDACs are responsible for repressing ARHI, we cotransfected vectors expressing HDACs 1-11 with an ARHI/luciferase reporter into SKBr3 and MCF-7 breast cancer cells. Expression of multiple HDACs consistently reduced ARHI promoter activity in a dose-dependent manner. We also found that the expression level of HDACs 1-3 was higher in breast cancer cell lines than in normal breast epithelial cells. In agreement with their repressive function, depletion of HDACs 1, 3 and 11 not only significantly increased the ARHI promoter activity of the transfected reporter but also activated the transcription of the endogenous ARHI gene. Furthermore, depletion or inhibition of HDACs by small interfering RNA of HDAC11 or by trichostatin A, respectively, increased E2F acetylation. Chromatin immunoprecipitation assays revealed that HDACs 1 and 3 are bound to the ARHI promoter. Taken together, our results suggest that the activity of multiple HDACs contributes to the repression of the ARHI tumor suppressor gene in breast cancer cells. Since HDAC inhibitors are now being used to treat breast cancer, the reactivation of ARHI in these cancer cells may serve as a new biomarker with which to monitor the treatment effects.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor/fisiologia , Inibidores de Histona Desacetilases , Proteínas rho de Ligação ao GTP/genética , Acetilação , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular , Núcleo Celular/metabolismo , Imunoprecipitação da Cromatina , Metilação de DNA/efeitos dos fármacos , Fator de Transcrição E2F1/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Luciferases/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Proteínas rho de Ligação ao GTP/metabolismo
16.
Clin Cancer Res ; 12(8): 2404-13, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16638845

RESUMO

PURPOSE: ARHI expression is lost or markedly down-regulated in the majority of ovarian cancers. The mechanism by which ARHI is down-regulated in ovarian cancers is still not clear. Our previous reports indicated that ARHI promoter activity was reduced in ovarian cancer cells, due in part to the effects of negative regulatory transcription factor(s). EXPERIMENTAL DESIGN AND RESULTS: We now show that E2F1 and E2F4, but not E2F2, E2F3, or E2F5, bind to the ARHI promoter and repress its activity in ovarian cancer cells. Consistent with this observation, immunochemical staining of cell lines and of 364 samples of ovarian cancer tissue show that the expression of E2F1 and E2F4 proteins is much higher in ovarian cancer cells than in normal ovarian epithelial cells, and that increased expression of E2Fs was negatively correlated with ARHI expression (P < 0.05). Mutation of the putative E2F binding site in the ARHI promoter reversed this inhibitory effect and significantly increased ARHI promoter activity. In addition to the effects of transcriptional regulation, ARHI mRNA also exhibited a significantly reduced half-life in ovarian cancer cells when compared with that in normal ovarian epithelial cells (P < 0.01), suggesting posttranscriptional regulation of ARHI expression. ARHI mRNA contains AU-rich elements (ARE) in the 3'-untranslated region. We have found that these AREs interact with HuR, an ARE-binding protein that stabilizes bound mRNAs, possibly contributing to the rapid turnover of ARHI mRNA. Finally, reduced HuR ARE binding activity was observed in ovarian cancer cells when compared with normal ovarian surface epithelium. CONCLUSIONS: Taken together, our data suggest that ARHI expression is regulated at both the transcriptional and the posttranscriptional levels, contributing to the dramatic decrease in ARHI expression in ovarian cancers.


Assuntos
Genes Supressores de Tumor , Neoplasias Ovarianas/genética , Processamento Pós-Transcricional do RNA/genética , Transcrição Gênica/genética , Proteínas rho de Ligação ao GTP/genética , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular Tumoral , Células Cultivadas , Regulação para Baixo/genética , Fatores de Transcrição E2F/genética , Fatores de Transcrição E2F/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Regulação Neoplásica da Expressão Gênica , Impressão Genômica , Humanos , Luciferases/genética , Luciferases/metabolismo , Neoplasias Ovarianas/patologia , Regiões Promotoras Genéticas/genética , Ligação Proteica , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Cancer Res ; 65(15): 6701-10, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16061651

RESUMO

A Ras homologue member I (ARHI) is a novel imprinted tumor suppressor gene whose expression is frequently lost in breast and ovarian cancers. This small GTP-binding protein is a member of the Ras superfamily with significant homology to both Ras and Rap. Unlike the Ras oncogene, however, ARHI inhibits tumor cell growth. To elucidate the mechanisms by which ARHI inhibits cancer growth, we screened a human breast epithelial cell cDNA library using a yeast two-hybrid system for ARHI-interacting proteins. ARHI was found to interact with signal transducers and activators of transcription (STAT) 3, a latent transcription factor that transduces signals from the cell surface to the nucleus and activates gene transcription. STAT3 is frequently phosphorylated and activated in breast and ovarian cancers, where cytokines and growth factors up-regulate STAT3 and stimulate proliferation. The ARHI-STAT3 interaction was confirmed by coimmunoprecipitation in mammalian cells and shown to be specific for STAT3 but not STAT1 or STAT5a. When ARHI and STAT3 were coexpressed in SKOv3 cells, ARHI formed a complex with STAT3 in the cytoplasm and prevented interleukin-6-induced STAT3 accumulation in the nucleus. ARHI markedly reduced STAT3 binding to DNA and STAT3-dependent promoter activity while only moderately affecting STAT3 phosphorylation. Deletion of the NH2 terminus of ARHI significantly compromised its inhibitory activity, suggesting that this unique NH2-terminal extension contributes to ARHI's inhibition of STAT3-mediated transcriptional activity. Thus, the physical association between STAT3 and ARHI as well as the functional inhibition of STAT3 transcriptional activity by ARHI suggests a novel mechanism through which a putative tumor suppressor gene can inhibit STAT3 activity in breast and ovarian cancers.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Neoplasias Ovarianas/metabolismo , Transativadores/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/fisiologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Humanos , Neoplasias Ovarianas/genética , Fosforilação , Ligação Proteica , Fator de Transcrição STAT3 , Transdução de Sinais , Transativadores/biossíntese , Transativadores/genética , Transativadores/metabolismo , Ativação Transcricional , Transfecção , Proteínas rho de Ligação ao GTP/metabolismo
18.
Lab Invest ; 84(8): 941-51, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15184909

RESUMO

Loss of phosphatase and tensin homolog (PTEN) and amplification of the epidermal growth factor receptor (EGFR) gene contribute to the progression of gliomas. As downstream targets of the PTEN and EGFR signaling pathways, Akt, NFkappaB, and signal transducer and activator of transcription-3 (Stat3) have been shown to play important roles in the control of cell proliferation, apoptosis, and oncogenesis. We examined the activation status of Akt, NFkappaB, and Stat3 in 259 diffuse gliomas using tissue microarrays and immunohistochemistry, and evaluated their association with glioma grade. We observed significant positive correlations between the activation status of Akt and NFkappaB and glioma grade. In contrast, only focal immunoreactivity for phospho-Stat3 was observed in < 9% of high-grade gliomas. In addition, we observed a significant correlation between the activation of Akt and NFkappaB. Functional correlation between Akt activation and the activation of NFkappaB was confirmed in U251MG GBM cells in which inhibition of Akt activation either by stable expression of PTEN or by the PI3-kinase inhibitors, wortmannin and LY294002, led to a concomitant decrease in NFkappaB-binding activity. Thus, our results demonstrate that constitutive activation of Akt and NFkappaB, but not Stat3, contributes significantly to the progression of diffuse gliomas, and activation of Akt may lead to NFkappaB activation in high-grade gliomas.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Glioma/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Estudos de Casos e Controles , Proteínas de Ligação a DNA/genética , Ativação Enzimática , Expressão Gênica , Genes erbB-1 , Glioma/genética , Glioma/patologia , Humanos , Imuno-Histoquímica , NF-kappa B/genética , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Análise Serial de Proteínas , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Fator de Transcrição STAT3 , Transdução de Sinais , Transativadores/genética , Proteínas Supressoras de Tumor/genética
19.
Biochem Biophys Res Commun ; 316(4): 1022-8, 2004 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-15044087

RESUMO

Interleukin-1 (IL-1) binds to its type I receptors (IL-1R), which in complex with IL-1R accessory protein (IL-1R AcP) induces various intracellular signaling events. We report here that IL-1 triggers the recruitment of phosphoinositide 3-kinase (PI 3-kinase) to a signaling complex and induces its lipid kinase activity in a biphasic manner. This IL-1-induced complex consists of IL-1R, IL-1R AcP, PI 3-kinase, and the IL-1-receptor-associated kinase (IRAK). Deletion of the C-terminus 27 amino acids of IL-1R AcP resulted in a mutant, CDelta27, that could not recruit PI 3-kinase to the signalsome nor stimulate PI3-kinase activity. Moreover, CDelta27 functioned as a dominant-negative mutant that inhibited IL-1-induced PI 3-kinase and NFkappaB activation. CDelta27, however, had no effect on IL-1-dependent activation of the Jun N-terminal kinase (JNK), indicating that distinct regions of IL-1R AcP mediate the activation of PI 3-kinase and JNK. Thus, our results identified a functional region in the IL-1R AcP required for the recruitment and activation of PI 3-kinase.


Assuntos
Carcinoma Hepatocelular/metabolismo , Carcinoma/metabolismo , Rim/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas/química , Proteínas/metabolismo , Receptores de Interleucina-1/metabolismo , Transdução de Sinais , Linhagem Celular , Ativação Enzimática , Humanos , Subunidade alfa de Receptor de Interleucina-18 , Proteínas Quinases JNK Ativadas por Mitógeno , Rim/embriologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Fosfatidilinositol 3-Quinases/química , Ligação Proteica , Receptores de Interleucina-1/química , Receptores Tipo I de Interleucina-1 , Receptores de Interleucina-18 , Relação Estrutura-Atividade
20.
J Biol Chem ; 279(9): 7576-83, 2004 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-14662759

RESUMO

Many cancers have constitutively activated NFkappaB, the elevation of which contributes to cancer cell resistance to chemotherapeutic agent-induced apoptosis. Although mitogen-activated protein kinase/extracellular-regulated kinase kinase kinase-3 (MEKK3) has been shown to participate in the activation of NFkappaB, its relations to apoptosis and cancer are unclear. In this study, we established cell model systems to examine whether stable expression of MEKK3 could lead to increased NFkappaB activity and confer resistance to apoptosis. In addition, we investigated in breast and ovarian cancers whether MEKK3 expression may be altered and correlated with aberrant NFkappaB activity. We show that stable cell lines overexpressing MEKK3 not only had elevated levels of NFkappaB binding activity but also were more responsive to cytokine stimulation. These stable cells showed 2-4-fold higher basal expression of Bcl-2 and xIAP than the parental cells. Consistent with this increased expression of cell survival genes, MEKK3 stable cells showed reduced activation of caspases 3 and 8 and poly(ADP-ribose) polymerase cleavage and dramatically increased resistance to apoptosis induced by tumor necrosis factor-related apoptosis-inducing ligand, doxorubicin, daunorubicin, camptothecin, and paclitaxel. Intriguingly, analysis of human breast and ovarian cancers showed that a significant fraction of these samples have elevated MEKK3 protein levels with corresponding increases in NFkappaB binding activities. Thus, our results established that elevated expression of MEKK3 appears to be a frequent occurrence in breast and ovarian cancers and that overexpression of MEKK3 in cells leads to increased NFkappaB activity and increased expression of cell survival factors and ultimately contributes to their resistance to apoptosis. As such, MEKK3 may serve as a therapeutic target to control cancer cell resistance to cytokine- or drug-induced apoptosis.


Assuntos
Apoptose , Expressão Gênica , MAP Quinase Quinase Quinases/genética , NF-kappa B/fisiologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Neoplasias da Mama/química , Neoplasias da Mama/metabolismo , Carcinoma Hepatocelular , Linhagem Celular , Sobrevivência Celular , Resistência a Medicamentos , Resistencia a Medicamentos Antineoplásicos , Embrião de Mamíferos , Feminino , Glioma , Humanos , Interleucina-1/farmacologia , Rim , Neoplasias Hepáticas , Luciferases/genética , MAP Quinase Quinase Quinase 3 , MAP Quinase Quinase Quinases/análise , MAP Quinase Quinase Quinases/fisiologia , Glicoproteínas de Membrana/farmacologia , NF-kappa B/análise , Neoplasias Ovarianas/química , Neoplasias Ovarianas/metabolismo , Proteínas/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Recombinantes de Fusão , Ligante Indutor de Apoptose Relacionado a TNF , Transfecção , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA