Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
2.
Am J Physiol Regul Integr Comp Physiol ; 302(9): R1084-90, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22338080

RESUMO

The opioid growth factor (OGF) and its receptor, OGFr, play a regulatory role in cell proliferation, and maintain homeostasis through a tonically active negative feedback mechanism. To directly evaluate the repercussion of increased OGFr expression and consequent gain-of-function in epithelium, bovine keratin 5 promoter elements were used to direct the expression of OGFr to skin in a tetracycline-regulated manner. Three founder lines overexpressing OGFr (OGFrTG/K5-tTA) were established. Evidence for increased OGFr in the epithelium included a three-fold increase in OGFr binding activity, as well as significant increases in OGFr protein, as monitored by semiquantitative immunohistochemistry. DNA synthesis in target epithelium, including cornea, tongue, and skin of transgenic mice was decreased 41% to 80% from wild-type littermates; the liver, a nonepithelial organ, was not altered. Decreased DNA synthesis in corneal epithelium induced by transgenic expression of OGFr was further reduced by treatment with exogenous OGF but reversed by exposure to the opioid antagonist, naloxone. The number of cell layers in both epidermis and cornea of OGFrTG/K5-tTA animals was reduced nearly 45% from wild-type mice. Full-thickness wounds in mice overexpressing OGFr healed 37% to 75% slower than wild-type littermates. These data demonstrate for the first time that stable genetic amplification of OGFr downregulates homeostatic cell proliferation, as well as pathophysiological processes with respect to wound repair. These mice also can serve as a valuable model to dissect the mechanism of OGF-OGFr action and may be important in understanding the etiology, pathogenesis, and treatment of epithelium-related diseases.


Assuntos
Epitélio/fisiologia , Peptídeos Opioides/metabolismo , Receptores Opioides/metabolismo , Fenômenos Fisiológicos da Pele , Cicatrização/fisiologia , Animais , Camundongos , Camundongos Transgênicos , Regulação para Cima
3.
Exp Biol Med (Maywood) ; 234(5): 532-41, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19244545

RESUMO

The opioid growth factor receptor (OGFr) mediates the inhibitory action of OGF on cell replication of normal and neoplastic cells. The spatiotemporal course of OGFr nucleocytoplasmic trafficking was determined with a probe of full-length OGFr fused to enhanced green fluorescent protein (eGFP). Translation of OGFr required 8.5 hours, and transit into the nucleus required 8 hours; OGFr remained in the nucleus for 8 days. OGFr was initially expressed on the outer nuclear envelope, transited to the paranuclear cytoplasm, and into the nucleus. Transport through the nuclear pore was elucidated by mutation of the nuclear localization signal (NLS) sequences in full-length OGFr. Mutation of each NLS reduced nuclear localization by 5%-50%, whereas simultaneous mutation of NLS383-386 and NLS456-460 abolished OGFr-eGFP nuclear localization in 80% of the cells. To determine whether intact NLSs are important for the inhibition of cell proliferation, DNA synthesis was monitored with BrdU. Wild-type OGFr-eGFP-transfected cells had 20% BrdU-positive cells, whereas cells with simultaneous mutation of all three NLS sites had a 70% labeling index. These results indicate that the regulation of cell proliferation by the OGF-OGFr axis is dependent on nucleocytoplasmic translocation and reliant on the integrity of two NLSs in OGFr to interact with transport receptors.


Assuntos
Proliferação de Células , DNA/biossíntese , Membrana Nuclear/metabolismo , Sinais de Localização Nuclear/metabolismo , Biossíntese de Proteínas/fisiologia , Receptores Opioides/biossíntese , Transporte Ativo do Núcleo Celular/fisiologia , Linhagem Celular Tumoral , DNA/genética , Humanos , Mutação , Membrana Nuclear/genética , Sinais de Localização Nuclear/genética , Receptores Opioides/genética , Fatores de Tempo
4.
Mol Cancer ; 7: 5, 2008 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-18190706

RESUMO

BACKGROUND: Pancreatic cancer is the 4th leading cause of death from cancer in the U.S. The opioid growth factor (OGF; [Met5]-enkephalin) and the OGF receptor form an inhibitory growth regulatory system involved in the pathogenesis and treatment of pancreatic cancer. The OGF-OGFr axis influences the G0/G1 phase of the cell cycle. In this investigation, we elucidate the pathway of OGF in the cell cycle. RESULTS: Using BxPC-3 cells, OGF decreased phosphorylation of retinoblastoma (Rb) protein without changing total Rb. This change was correlated with reduced cyclin-dependent kinase protein (Cdk) 2 kinase activity, but not total Cdk2. OGF treatment increased cyclin-dependent kinase inhibitor (CKI) p21 protein expression in comparison to controls, as well levels of p21 complexed with Cdk2. Naloxone abolished the increased expression of p21 protein by OGF, suggesting a receptor-mediated activity. p21 specific siRNAs blocked OGF's repressive action on proliferation in BxPC-3, PANC-1, and Capan-2 cells; cells transfected with negative control siRNA had no alteration in p21 expression, and therefore were inhibited by OGF. CONCLUSION: These data are the first to reveal that the target of cell proliferative inhibitory action of OGF in human pancreatic cancer is a p21 CKI pathway, expanding strategies for diagnosis and treatment of these neoplasias.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Encefalina Metionina/análogos & derivados , Neoplasias Pancreáticas/metabolismo , Receptores Opioides/metabolismo , Transdução de Sinais/fisiologia , Western Blotting , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Quinase 2 Dependente de Ciclina/metabolismo , Progressão da Doença , Encefalina Metionina/metabolismo , Citometria de Fluxo , Humanos , RNA Interferente Pequeno , Receptores Opioides/genética , Proteína do Retinoblastoma/metabolismo , Transfecção
5.
Breast Cancer Res Treat ; 112(2): 255-61, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18097746

RESUMO

We have shown that administration of alpha-difluoromethylornithine (DFMO), an irreversible inhibitor of ornithine decarboxylase (ODC), the first and rate-limiting enzyme in polyamine (PA) biosynthesis, reduces the invasive and metastatic properties of MDA-MB-435 breast cancer cells while activating multiple signal transduction pathways, including MAPK, Stat3, Stat1, and JNK. Since the activity of these signaling mechanisms is frequently regulated by upstream tyrosine kinases (TKs), we tested whether non-receptor and receptor TKs may be involved in the signaling and biological effects of DFMO in MDA-MB-435 cells. Treatment with DFMO (1 mM for 48 h) did not affect Src phosphorylation (Tyr 416). Administration of the Src-family members inhibitor PP-1 (1 microM), blocked Src phosphorylation in the absence and in the presence of DFMO, but did not block the signaling effects of DFMO (increased phosphorylation of Stat3, Stat1, ERK and JNK). PP-1 treatment, on the other hand, inhibited the invasiveness of MDA-MB-435 cells in matrigel and potentiated the anti-invasive effect of DFMO. Next, we focused on the role of receptor TK. Western analysis of cell lysates from MDA-MB-435 cells failed to show the presence of EGF-R and HER-2neu but demonstrated the expression of c-Met, the receptor for hepatocyte growth factor (HGF). Therefore, we tested the effect of DFMO on the HGF/c-Met pathway which is strongly implicated in the progression of human breast cancer. We found that DFMO treatment blocked HGF-induced c-Met phosphorylation in MDA-MB-435 cells, suggesting that its anti-invasion action may be mediated, at least in part, by blocking c-Met signaling. Next, we showed that 1 mM DFMO suppressed HGF induced invasiveness of MDA-MB-435 cells in matrigel. Combination administration of DFMO with suboptimal doses of PHA-665752, a specific c-Met inhibitor, reduced invasiveness to an even greater extent than the individual treatment. These findings indicate that Src-family members, while not involved in DFMO action, promote invasiveness of breast cancer cells and their inhibition may enhance the antitumor effect of PA depletion. Our data also point to inhibition of HGF/c-Met pathway as a possible novel approach to enhancing the antitumor action of DFMO.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Eflornitina/farmacologia , Proteínas Tirosina Quinases/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Antineoplásicos/farmacologia , Proliferação de Células , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Indóis/farmacologia , Invasividade Neoplásica , Fosforilação , Poliaminas/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais , Sulfonas/farmacologia , Quinases da Família src/metabolismo
6.
Breast Cancer Res Treat ; 107(1): 63-70, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17333334

RESUMO

We have shown that alpha-difluoromethylornithine (DFMO), an inhibitor of ornithine decarboxylase, the first and rate-limiting enzyme in polyamine synthesis, has significant antiproliferative and antiinvasive effects in breast cancer cells. We have also reported that these antitumor effects are associated with activation of multiple signaling pathways, including STAT-3, STAT-1, Jun-N-Terminal kinase (JNK), and Mitogen activated protein kinase (MAPK), the latter being found to mediate its antiinvasive action in MDA-MB-435 cells. The present experiments were designed to test the effect of DFMO on the protein kinase A (PKA) pathway and determine its biological significance. We found that DFMO administration (1 mM) to MDA-MB-435 breast cancer cells significantly increased cAMP response element (CRE)-binding protein (CREB) phosphorylation as well as the transactivation of pCRE-luc, a CREB-dependent promoter activated by PKA. To determine the significance of this biochemical effect of DFMO, we used the PKA inhibitor H89 which, as expected, suppressed in a dose-dependent manner (1 and 10 microM) basal and DFMO-induced CREB phosphorylation in our system. Administration of H89 alone was able to suppress proliferation of MDA-MB-435 cells when used at a concentration (10 microM) shown to completely block basal CREB phosphorylation. At concentrations of 0.5 and 1 muM, H89 treatment, while having no antiproliferative effect of its own, potentiated in a dose-dependent fashion the growth inhibitory action of a suboptimal concentration of DFMO (0.01 mM). Ten micromoles of H89 reduced invasiveness of MDA-MB-435 cells in matrigel by approximately 40% (an effect similar to that of 1 mM DFMO). The combination treatment further reduced invasiveness by approximately 80% (P < 0.01 versus the individual treatments). H89 treatment (10 microM) partially reduced DFMO-induced phosphorylation of STAT-3 but not that of STAT-1, Extracellular regulated kinase (ERK), and JNK. In conclusion, our results indicate that PKA signaling exerts proproliferative and proinvasive effects in our experimental system. Therefore, its activation by DFMO represents a compensatory mechanism which should be blocked in order to maximize the antitumor action of the drug. Our data are also consistent with the notion that STAT-3 activation by DFMO is at least in part mediated through the PKA pathway.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Eflornitina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Isoquinolinas/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica , Fosforilação , Regiões Promotoras Genéticas , Transdução de Sinais , Sulfonamidas/farmacologia
7.
Breast Cancer Res Treat ; 107(1): 33-40, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17333337

RESUMO

Increased polyamine synthesis has been associated with proliferation and progression of breast cancer, and thus, is a potential target for anticancer therapy. Polyamine depletion by alpha-difluoromethylornithine (DFMO) has been shown to decrease pulmonary and bone metastasis from human breast cancer cell xenografts. Following these observations, this study was designed to test the effects of DFMO on in vitro and in vivo features of the highly invasive and metastatic 4T1 murine mammary cancer cells. DFMO inhibited proliferation, caused G1-S arrest, and suppressed in vitro invasiveness of 4T1 cells. In contrast to our previous findings with MDA-MB-435 cells, DFMO did not affect the activation of signal transducers and activator of transcription 3, c-Jun N-terminal kinase, and extracellular signal-regulated kinase, but decreased phosphorylation of p38. DFMO did not alter the expression of Twist. DFMO delayed the orthotopic growth of 4T1 xenografts in association with suppressed putrescine and spermidine levels but increased levels of spermine. DFMO did not affect pulmonary metastasis when primary tumors of control and DFMO-treated mice were matched for size. Interestingly, DFMO reduced Ki-67 expression only in the primary tumors but did not affect its expression in the metastatic tumors in the lung. Cleaved caspase-3 expression was not affected by DFMO in either the primary tumors or the pulmonary metastasis. In summary, DFMO treatment markedly inhibited in vitro proliferation and invasiveness of 4T1 cells and retarded the growth of orthotopic xenografts in mice. The failure of DFMO to inhibit pulmonary metastasis in this system appears to be due, at least in part, to its lack of antiproliferative effect at the metastatic sites.


Assuntos
Eflornitina/metabolismo , Neoplasias Mamárias Animais/metabolismo , Poliaminas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Técnicas In Vitro , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Pulmonares/metabolismo , Camundongos , Metástase Neoplásica , Transplante de Neoplasias , Fosforilação , Fator de Transcrição STAT3/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Elife ; 72018 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-29848440

RESUMO

The training of PhD students and early-career scientists is largely an apprenticeship in which the trainee associates with an expert to become an independent scientist. But when is a PhD student ready to graduate, a postdoctoral scholar ready for an independent position, or an early-career scientist ready for advanced responsibilities? Research training by apprenticeship does not uniformly include a framework to assess if the trainee is equipped with the complex knowledge, skills and attitudes required to be a successful scientist in the 21st century. To address this problem, we propose competency-based assessment throughout the continuum of training to evaluate more objectively the development of PhD students and early-career scientists.


Assuntos
Escolha da Profissão , Educação de Pós-Graduação , Avaliação Educacional , Competência Mental , Pesquisadores , Estudantes , Humanos , Pensamento
9.
Mol Cell Endocrinol ; 278(1-2): 29-35, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17888567

RESUMO

We have established the gene for IGF binding protein-3 (IGFBP-3) as a target for FSH action. FSH effects on this gene require the PKA pathway as well as the PI-3 kinase and MAPK pathways. At the IGFBP-3 promoter, FSH effects depend on a site for TATA box binding protein (TBP) and formation of a high molecular weight transcription complex. To further elucidate FSH effects on the downstream events involving the TBP site, we cloned a pig TAF4b cDNA into a P-Flag expression vector. By co-transfecting granulosa cells with the IGFBP-3 promoter, we found that TAF4b mimics and enhances FSH induction of IGFBP-3 reporter activity. Using RT-PCR we showed that FSH stimulates expression of TAF4b. This would suggest that the role of TAF4b in follicular development is regulated by FSH. TAF4b may thus be the TFIID component that binds to the TBP site on the IGFBP-3 promoter and is essential for FSH induction of IGFBP-3.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Células da Granulosa/metabolismo , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Ovário/metabolismo , Sus scrofa/metabolismo , Fator de Transcrição TFIID/fisiologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Clonagem Molecular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Hormônio Foliculoestimulante/farmacologia , Regulação da Expressão Gênica , Genes Reporter/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , Ovário/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-raf/metabolismo , RNA Mensageiro/metabolismo , Sus scrofa/genética , Fator de Transcrição TFIID/genética , Transcrição Gênica/efeitos dos fármacos
10.
Int J Oncol ; 30(4): 775-83, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17332915

RESUMO

The opioid growth factor (OGF), [Met5]-enkephalin, and OGF receptor (OGFr) form an inhibitory axis regulating the growth of human pancreatic cancer. This study examined whether overexpression of OGFr decreases the growth of pancreatic cells in vitro. MIA PaCa-2 cells were transfected with OGFr cDNA, and six clonal lines were examined for protein expression and function. OGFr binding assays revealed a 2.3- to 5.6-fold increase in binding capacity from wild-type (WT) and empty vector (EV) controls; binding affinity was comparable in all groups. OGFr protein expression, as measured by immunohistochemistry and Western blotting, was enhanced in clonal cell lines compared to controls. Doubling times of OGFr clonal lines were 47-91% longer than in the WT/EV groups for all but one clonal line. DNA synthesis of cells overexpressing OGFr was diminished from the WT/EV groups by 28-52%. Addition of exogenous OGF further reduced (14-31%) the cell growth of clonal lines, and the effects of exogenous OGF were receptor-mediated. Exposure of cells overexpressing OGFr to naltrexone increased the cell number by up to 9.4-fold. OGF was identified as the only opioid peptide to depress cell replication in the transfected cell lines. Neutralization of endogenous OGF with antibodies to this peptide elevated the cell number in clonal cell lines. These data identify OGFr at the molecular level as integral to regulating the cell replication of human pancreatic cancer, and support treatment modalities that amplify OGFr in order to decrease the growth of these neoplasias.


Assuntos
Proliferação de Células , Neoplasias Pancreáticas/patologia , Receptores Opioides/metabolismo , Contagem de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA/biossíntese , Terapia Genética , Humanos , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/terapia , Receptores Opioides/análise , Receptores Opioides/genética , Transfecção , Regulação para Cima
11.
Int J Oncol ; 31(5): 1187-91, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17912446

RESUMO

We have previously observed that inhibition of polyamine biosynthesis with alpha-difluoromethylornithine (DFMO) upregulates production of thrombospondin-1 (TSP-1), an extracellular matrix protein with potent anti-angiogenic and antimetastatic properties, by MDA-MB-435 human breast cancer cells in culture. The present experiments were designed to investigate the mechanisms by which DFMO regulates TSP-1 production in this system. 35S-methionine pulse chase experiments indicated that DFMO administration increased TSP-1 synthesis by approximately 6-fold, while it slightly but significantly decreased protein half-life from 35 to 28 min. DFMO treatment increased steady state TSP-1 mRNA levels by 2-fold in MDA-MB-435 cells. TSP-1 promoter reporter studies indicated that this increase was largely due to activation of transcription. Analysis of distribution of TSP-1 mRNA levels between non-polysomal, subpolysomal and polysomal fractions in control and DFMO-treated cells suggested a major stimulatory effect of the drug on TSP-1 translation. A similar increase in TSP-1 transcription and translation in response to DFMO treatment was also observed in vivo in MDA-MB-435 breast cancer xenografts. Surprisingly however, we failed to detect an increase in TSP-1 protein as assessed by Western blot analysis. The reason for this unexpected finding is unknown but may be due to DFMO-induced stimulation of TSP-1 secretion into the systemic circulation, thus preventing its accumulation within the tumor.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Eflornitina/farmacologia , Inibidores Enzimáticos/farmacologia , Trombospondina 1/biossíntese , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , RNA Mensageiro/análise , Trombospondina 1/genética
12.
Int J Mol Med ; 19(3): 421-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17273790

RESUMO

The opioid growth factor (OGF) is a constitutively expressed negative growth regulator whose action is mediated by the OGF receptor (OGFr). The OGF-OGFr axis tonically regulates the growth of human squamous cell carcinoma of the head and neck (SCCHN). To examine the repercussions of amplifying OGFr in SCCHN, constructs were prepared to overexpress OGFr in SCC-1 cells; six clonal lines were examined. OGFr binding assays of clonal cells revealed a 2.4- to 8.4-fold increase in binding capacity compared to wild-type (WT) and empty vector (EV) controls; binding affinity was comparable in all groups. OGFr protein expression, as measured by quantitative immunohistochemistry and Western blotting, was increased in clonal cell lines compared to controls. Under standard growth conditions the cell number of the OGFr clonal lines was reduced by 11 to 68% from the WT group, and doubling times were 7 to 67% longer. Addition of exogenous OGF further reduced (8 to 37%) cell growth of the clonal lines. Depletion of endogenous OGF with antibodies to this peptide increased growth 2-fold in cells amplifying OGFr relative to increases of 32 and 34% for the WT and EV groups, respectively. DNA synthesis of cells overexpressing OGFr was reduced from the WT group by 46 to 75%. These data indicate that the OGF receptor is integral to cell replication of SCCHN, and support treatment modalities that amplify OGFr in order to decrease the growth of these neoplasias.


Assuntos
Carcinoma de Células Escamosas/patologia , Regulação para Baixo , Expressão Gênica , Neoplasias de Cabeça e Pescoço/patologia , Receptores Opioides/metabolismo , Idoso , Anticorpos/farmacologia , Bromodesoxiuridina/metabolismo , Contagem de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Clonais , DNA de Neoplasias/biossíntese , Regulação para Baixo/efeitos dos fármacos , Encefalina Metionina/imunologia , Encefalina Metionina/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Masculino , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Receptores Opioides/genética
13.
Cancer Res ; 65(23): 11026-33, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16322252

RESUMO

Inhibition of polyamine biosynthesis with alpha-difluoromethylornithine (DFMO) has been shown to inhibit proliferation of breast cancer cells although its mechanism of action has not been fully elucidated. To address this issue, we tested the effects of DFMO on cell cycle variables of MDA-MB-435 human breast cancer cells in culture. We also focused on the possible mediatory role of the mitogen-activated protein kinase (MAPK) pathway on the cell cycle effects of DFMO because this compound has been shown to activate MAPK signaling. We found that DFMO caused a p53-independent increase in p21 and its association with cyclin-dependent kinase (cdk)-2 and decreased cdk-2 protein as well as its phosphorylation on Thr160. In addition, DFMO markedly suppressed the expression of the full-length and low molecular weight forms of cyclin E. These effects of DFMO were reversible with exogenous putrescine, thus indicating that they are specifically mediated through polyamine depletion. Cdk-2 activity was drastically reduced in DFMO-treated breast cancer cells which exhibited a reduction in retinoblastoma (Rb) phosphorylation and protein. As a predictable consequence of these effects, DFMO caused a G1-S block. In addition, DFMO inhibited G2-M transition, most likely as a result of its induction of p21 expression. Inhibition of the MAPK pathway with PD98059 or U0126 blocked the DFMO-induced induction of p21 and the reduction of cdk-2 protein. PD98059 reversed the G2-M block induced by DFMO (probably as a result of suppression of p21) but not the G1-S arrest. MDA-MB-435 cells treated with PD98059 or U0126 in the presence and absence of DFMO exhibited a marked increase in the expression of p27 and its association with cdk-2, a decrease in phosphorylation of cdk-2 on Thr160, and a decrease in cyclin E expression. As predicted, PD98059 treatment reduced cdk-2 activity and Rb phosphorylation while reversing the decrease in Rb protein induced by DFMO. Neither DFMO nor PD98059, either alone or in combination, reduced cdk-4 activity despite a marked induction in p15 expression caused by DFMO. Our results indicate that activation of the MAPK pathway accounts for some of the effects of DFMO on cell cycle events of breast cancer cells. Inhibition of the MAPK pathway, however, does not reverse the cell cycle arrest induced by DFMO because of activation of alternative mechanisms leading to suppression of cdk-2 activity.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Eflornitina/farmacologia , Sistema de Sinalização das MAP Quinases/fisiologia , Putrescina/farmacologia , Butadienos/farmacologia , Proteínas de Ligação ao Cálcio/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Quinase 2 Dependente de Ciclina/biossíntese , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Interações Medicamentosas , Flavonoides/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neoplasias Hormônio-Dependentes/enzimologia , Neoplasias Hormônio-Dependentes/patologia , Nitrilas/farmacologia , Fosforilação , Proteína do Retinoblastoma/metabolismo
14.
Mol Endocrinol ; 19(7): 1837-48, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15718291

RESUMO

The current study was done to elucidate the mechanism of the FSH stimulation of IGF-binding protein 3 (IGFBP-3) expression and map the FSH response element on the pig IGFBP-3 promoter. Forskolin induced IGFBP-3 reporter activity in transiently transfected granulosa cells. The protein kinase A (PKA) inhibitor [N-[2-(p-bromocinnamyl)amino)ethyl]-5-isoquinolinesulfonamide, 2HCl] (and cotransfection with a PKA inhibitor expression vector), the phosphatidylinositol-3 kinase inhibitor [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one], and the ERK inhibitor [1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)butadiene], all blocked FSH stimulation. Use of serial deletion constructs and site-directed mutagenesis show that a TATA box-binding protein site is required for FSH stimulation and that a specific protein 1 (Sp1) site is required for basal transcription. Gel shift assays of nuclear protein with a -61/-25 probe detected four protein-DNA complexes, with bands I and II having significantly higher intensities in FSH-treated cells than in controls. Mutation of the Sp1 site prevented formation of bands I and II whereas mutation of the TATA box-binding protein site prevented formation of band IV. Use of specific antibodies showed that Sp1 participates in formation of band I, Sp3 band II, and p300 in both I and II. Band III was nonspecifically competed out. We conclude that FSH stimulation of IGFBP-3 transcription is mediated by cAMP via the PKA pathway and requires the P1-3 kinase and likely the MAPK pathways.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Células da Granulosa/metabolismo , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Transdução de Sinais , Proteína de Ligação a TATA-Box/metabolismo , Ativação Transcricional , Animais , Colforsina/farmacologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/enzimologia , Mutagênese Sítio-Dirigida , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Regiões Promotoras Genéticas/genética , Elementos de Resposta , Deleção de Sequência , Suínos , TATA Box/genética , Fatores de Transcrição/metabolismo
15.
Clin Exp Metastasis ; 22(3): 255-63, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16158253

RESUMO

We have previously reported that inhibition of polyamine biosynthesis with alpha-difluoromethylornithine (DFMO) reduces pulmonary metastasis from MDA-MB-435 human breast cancer xenografts without affecting the volume of the primary tumors (Manni et al. Clin Exp Mets 20:321, 2003). In these experiments, we show that DFMO treatment (2% in drinking H(2)O) reduced the growth fraction of the primary tumors by 60%. However, this effect was counter-balanced by a similar reduction in non-apoptotic necrosis, thus accounting for the preservation of tumor volume in DFMO-treated mice. DFMO treatment caused a 4-fold increase in cytoplasmic staining for cleaved caspase-3 (as opposed to the nuclear staining observed in control tonsil tissue) in the absence of histologic evidence of apoptosis. DFMO treatment reduced the number of mice with pulmonary metastasis by approximately 80% and the number of metastasis per mouse by >90% in association with a reduction in invasiveness of the primary tumor in the surrounding dermis and muscle by approximately 30%. DFMO treatment increased ERK phosphorylation in the tumors, an effect that has been found by us in vitro to be causally linked to the anti-invasive effect of the drug (Manni et al. Clin Exp Metast 2004; 21: 461]. DFMO also increased tyrosine phosphorylation of STAT-3 and expression of STAT-1 and JNK proteins. Administration of SAM486A (1 mg/kg/i.p. daily), an inhibitor of S-adenosylmethionine decarboxylase, either individually or in combination with DFMO, was not found to exert any biological or biochemical effects, most likely as a result of its failure to suppress tissue polyamine levels under these experimental conditions.


Assuntos
Neoplasias da Mama/metabolismo , Eflornitina/farmacologia , Neoplasias Pulmonares/metabolismo , Metástase Neoplásica/prevenção & controle , Poliaminas/metabolismo , Amidinas/administração & dosagem , Amidinas/farmacologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/secundário , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Eflornitina/administração & dosagem , Feminino , Humanos , Indanos/administração & dosagem , Indanos/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias , Poliaminas/química , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo
16.
Int J Oncol ; 26(3): 809-16, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15703840

RESUMO

This study evaluated the effects of a combination of opioid growth factor (OGF) and paclitaxel on squamous cell carcinoma of the head and neck (SCCHN) using a tissue culture model of human SCCHN. The combination of OGF and paclitaxel was markedly inhibitory to SCCHN proliferation, reducing growth from control levels by 48 to 69% within 48 h. OGF in combination with carboplatin also depressed cell growth. The effect of a combination of OGF and paclitaxel or carboplatin on SCCHN growth was greater than either of the individual compounds. The efficacy of OGF, but not paclitaxel, was mediated by a naloxone-sensitive receptor and completely reversible. OGF, but no other endogenous or exogenous opioid, altered replication of SCCHN. OGF and paclitaxel depressed DNA synthesis, whereas only paclitaxel induced apoptosis. The combination of OGF and paclitaxel also had a supra-additive effect on the growth of another SCCHN, CAL-27, indicating the ubiquity of the combined drug activity. These data suggest that the combination of a biotherapy (OGF) and chemotherapy (paclitaxel and carboplatin) may provide an enhanced antitumor effect with respect to SCCHN.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Escamosas/patologia , Encefalina Metionina/farmacologia , Neoplasias de Cabeça e Pescoço/patologia , Paclitaxel/farmacologia , Idoso , Antineoplásicos/farmacologia , Apoptose , Carboplatina/farmacologia , Proliferação de Células , Humanos , Masculino , Células Tumorais Cultivadas
17.
Cancer Chemother Pharmacol ; 56(5): 510-20, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15947928

RESUMO

Gemcitabine is the standard of care for advanced pancreatic neoplasia, and exerts its effect through inhibition of DNA synthesis. However, gemcitabine has limited survival benefits. Opioid growth factor (OGF) is an autocrine-produced peptide that interacts with the nuclear receptor, OGFr, to inhibit cell proliferation but is not cytotoxic or apoptotic. The present study was designed to examine whether a combination of chemotherapy with gemcitabine and biotherapy with OGF is more effective than either agent alone in inhibiting pancreatic cancer growth in vitro and in vivo. The combination of OGF (10(-6) M) and gemcitabine (10(-8) M) reduced MIA PaCa-2 cell number from control levels by 46% within 48 h, and resulted in a growth inhibition greater than that of the individual compounds. OGF in combination with 5-fluorouracil also depressed cell growth more than either agent alone. The action of OGF, but not gemcitabine, was mediated by a naloxone-sensitive receptor, and was completely reversible. OGF, but no other endogenous or exogenous opioids, altered pancreatic cancer growth in tissue culture. The combination of OGF and gemcitabine also repressed the growth of another pancreatic cancer cell line, PANC-1. MIA PaCa-2 cells transplanted into athymic mice received 10 mg/kg OGF daily, 120 mg/kg gemcitabine every 3 days; 10 mg/kg OGF daily and 120 mg/kg gemcitabine every 3rd day, or 0.1 ml of sterile saline daily. Tumor incidence, and latency times to tumor appearance, of mice receiving combined therapy with OGF and gemcitabine, were significantly decreased from those of the control, OGF, and gemcitabine groups. Tumor volumes in the OGF, gemcitabine, and OGF/gemcitabine groups were markedly decreased from controls beginning on days 14, 12, and 8, respectively, after tumor cell inoculation. Tumor weight and tumor volume were reduced from control levels by 36-85% in the OGF and/or gemcitabine groups on day 45 (date of termination), and the group of mice exposed to a combination of OGF and gemcitabine had decreases in tumor size of 70% and 63% from the OGF or the gemcitabine alone groups, respectively. This preclinical evidence shows that combined chemotherapy (e.g. gemcitabine) and biotherapy (OGF) provides an enhanced therapeutic benefit for pancreatic cancer.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antimetabólitos Antineoplásicos/uso terapêutico , Desoxicitidina/análogos & derivados , Encefalina Metionina/uso terapêutico , Substâncias de Crescimento/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Caspase 3 , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/administração & dosagem , Desoxicitidina/uso terapêutico , Quimioterapia Combinada , Encefalina Metionina/administração & dosagem , Fluoruracila/farmacologia , Substâncias de Crescimento/administração & dosagem , Substâncias de Crescimento/sangue , Humanos , Masculino , Camundongos , Camundongos Nus , Naloxona/farmacologia , Entorpecentes/farmacologia , Neoplasias Pancreáticas/patologia , Receptores Opioides/agonistas , Receptores Opioides/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
18.
Cancer Chemother Pharmacol ; 56(1): 97-104, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15791460

RESUMO

Paclitaxel is used as a single agent, and in combination with other drugs, as a standard of care in the treatment of squamous cell carcinoma of the head and neck (SCCHN). However, the use of paclitaxel for therapy of SCCHN may be accompanied by serious side effects. Paclitaxel is a known cytotoxic inhibitor of cell proliferation that acts by stabilizing microtubules and inducing apoptosis. Opioid growth factor (OGF), [Met(5)]-enkephalin, is an endogenous peptide that has tonically active inhibitory effects on the growth of SCCHN in vitro and in vivo. OGF action is rapid, reversible, mediated by the nuclear-associated OGF receptor (OGFr), and is not cytotoxic (nor apoptotic related). The present study was designed to examine whether a combination of chemotherapy with paclitaxel and biotherapy with OGF is more effective than either agent alone in inhibiting tumor growth. Moreover, focus was placed on whether there are changes in the side effects known to occur with paclitaxel alone, following this combined therapy. Human SCC-1 cells, derived from a well differentiated SCCHN, were transplanted into athymic mice. The mice were randomized to receive intraperitoneal (i.p.) injections of sterile saline (controls), OGF (10 mg/kg, daily), paclitaxel (8 mg/kg, every other day), or both paclitaxel (8 mg/kg, every other day) and OGF (10 mg/kg, daily) beginning on the day of tumor inoculation. OGF, but not paclitaxel, delayed measurable and visible tumor appearance of mice with SCCHN. Treatment with paclitaxel, but not with other agents, had a marked effect on the body weights. Survival only was reduced in the paclitaxel group, with an average life span of 34.3+/-3.1 days recorded, in comparison to the 50-day survival (date of termination) for all other groups. Beginning after week 4 of tumor inoculation and drug treatment, the tumor weight of the paclitaxel/OGF group was significantly reduced from the control, OGF, and paclitaxel-exposed mice. The OGFr number of the SCCHN tumors was 2.1-fold greater in the animals exposed to OGF or paclitaxel, and elevated 38% in the paclitaxel/OGF group; significant differences from the control group were found for the OGF and paclitaxel groups. These data suggest that combined chemotherapy (i.e., paclitaxel) and biotherapy (OGF) provides a valuable alternative to the standard of care for SCCHN patients.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Escamosas/patologia , Encefalina Metionina/farmacologia , Neoplasias de Cabeça e Pescoço/patologia , Paclitaxel/farmacologia , Animais , Antineoplásicos Fitogênicos/efeitos adversos , Peso Corporal , Carcinoma de Células Escamosas/veterinária , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Neoplasias de Cabeça e Pescoço/veterinária , Masculino , Camundongos , Camundongos Nus , Paclitaxel/efeitos adversos , Análise de Sobrevida
19.
Cornea ; 24(5): 614-9, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15968171

RESUMO

PURPOSE: This study was designed to determine at the molecular level whether interactions between the opioid growth factor (OGF) and OGF receptor (OGFr) play a role in regulating DNA synthesis in the homeostasis of the corneal epithelium. METHODS: The plasmid pcDNA3.1+OGFr-HA, carrying the rat OGFr cDNA epitope-tagged with a C-terminal hemagglutinin (HA), or the empty-vector (pcDNA3.1+), was delivered twice by the Helios Gene Gun System at 300 psi to the cornea of anesthetized rats. The contralateral (untreated) cornea served as the naive specimen. BrdU was used to determine whether the recombinant OGFr was effective in regulating DNA synthesis in the rat peripheral corneal epithelium. RESULTS: Within 18 hours of transfection, positive HA staining was apparent in both the basal and suprabasal layers (efficiency > 90% of the cells) throughout the central and peripheral cornea. Quantitative immunohistochemistry with rhodamine-conjugated anti-OGFr antibodies revealed twofold more OGFr expression in the central and peripheral epithelium of transfected corneas relative to naive corneas. The number of BrdU-positive basal cells in the peripheral epithelium of the transfected cornea was one-third of that in the naive cornea. CONCLUSIONS: These data demonstrate the direct role of the OGF-OGFr system in determining cellular renewal in the mammalian corneal epithelium. Moreover, the successful establishment of a novel delivery system of cDNAs to the ocular surface suggests a therapeutic role for gene therapy in the eye.


Assuntos
Biolística/métodos , Proliferação de Células , DNA Complementar/genética , Epitélio Corneano/citologia , Receptores Opioides/fisiologia , Animais , Bromodesoxiuridina/metabolismo , Replicação do DNA , Encefalina Metionina/fisiologia , Técnica Indireta de Fluorescência para Anticorpo , Expressão Gênica , Vetores Genéticos , Masculino , Plasmídeos/genética , Ratos
20.
Endocrinology ; 145(4): 1776-85, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14715717

RESUMO

The role and regulation of IGF-binding protein-3 (IGFBP-3) in the ovary is not fully understood. We cloned and determined the sequence of 12,257 bp of the pig IGFBP-3 gene that includes 4,296 bp of the flanking promoter sequence. The porcine IGFBP-3 promoter sequence shares two highly conserved regions with the human and bovine IGFBP-3 promoters and a mouse DNA clone. The first is a 38 bp region between -1095 and -1058, whereas the second is a 73-bp region between -63 and +10 of the pig sequence. Projected translation of the open reading frame of our sequence gave a peptide sequence identical to that determined by peptide sequencing, but with 27 additional amino acids upstream of this sequence and is highly similar to the human, bovine, rat, and mouse IGFBP-3 peptides. Using RT-PCR we demonstrated that FSH regulates IGFBP-3 mRNA expression in a biphasic manner, with an early induction (maximal at 3 h) and an inhibition at 24 h after FSH treatment. The inhibition at 24 h was not due to changes in IGFBP-3 mRNA stability. A similar pattern of FSH modulation of the IGFBP-3 gene transcription was demonstrated by the reporter activity of granulosa cells transiently transfected with IGFBP-3 promoter constructs. The site for FSH stimulation of the IGFBP-3 gene was localized to the sequence between -61 and -48 relative to the transcription start site. Regulation of IGFBP-3 transcription by FSH suggests a role for IGFBP-3 in follicular development that may be independent of IGF-I.


Assuntos
Mapeamento Cromossômico , Clonagem Molecular , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/metabolismo , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Regiões Promotoras Genéticas/genética , Animais , Sequência de Bases , Feminino , Dados de Sequência Molecular , Homologia de Sequência do Ácido Nucleico , Suínos , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA