Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Physiol Biochem ; 43(6): 2310-2326, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29073599

RESUMO

BACKGROUND/AIMS: Solamargine, one natural photochemical component from traditional plants, has been shown to have anti-cancers properties. We previously showed that solamargine inhibited the growth of non-small-cell lung cancer (NSCLC) cells through suppression of prostaglandin E2 (PGE2) receptor EP4 gene and regulation of downstream signaling pathways. However, the detailed mechanism underlying this, especially in combination of metformin, a known AMPK activator, still remained to be determined. METHODS: Cell viability was measured using a 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) and colorimetric 5-bromo-2-deoxyuridine (BrdU) ELISA methods, respectively. Western blot analysis and immunohistochemistry were performed to examine the phosphorylation and protein expressions of signal transducer and activator of transcription 3 (Stat3), SP1, forkhead box O3a (FOXO3a), and insulin-like growth factor (IGF)-IGF binding protein 1 (IGFBP1). The expression of IGFBP1 mRNA was measured by quantitative real time PCR (qRT-PCR). Silencing of FOXO3a and IGFBP1 were examined by siRNA procedures. Exogenously expression of SP1, FOXO3a, and IGFBP1 were carried out by transient transfection assays. The promoter activity of IGFBP1 was tested using Secrete-PairTM Dual Luminescence Assay Kit. A xenografted tumor model was used to further test the effect of solamargine in combining with metformin in vivo. RESULTS: We further demonstrated that solamargine inhibited growth and induced cell cycle arrest in other NSCLC cell lines. Through mechanism-based approaches, we showed that solamargine decreased the phosphorylation of Stat3; In addition, solamargine induced FOXO3a, whereas reduced SP1 protein levels; all of which were abrogated in cells with overexpressed Stat3 gene. Interestingly, there is interaction between FOXO3a and SP1. Moreover, solamargine increased mRNA, protein expression and promoter activity of IGFBP1, which was not observed in cells with overexpressed SP1 or with silenced FOXO3a genes. Finally, ablation of IGFBP1 expression by siRNA blocked the effect of solamargine on cell growth inhibition. More importantly, there was a synergy of combination of solamargine and metformin. Similar findings were also observed in vivo. CONCLUSION: Our results show that solamargine increases IGFBP1 gene expression through inactivation of Stat3, followed by regulation and reciprocal interaction of FOXO3a and SP1 in vitro and in vivo. This ultimately leads to suppression of human lung cancer cell growth. Moreover, this is a synergy of solamargine in combination with metformin in this process. This study unravels a novel mechanism underlying the anti-lung cancer effects of solamargine in combination of metformin, and suggests a potential new lung cancer associated therapy.


Assuntos
Proteína Forkhead Box O3/metabolismo , Expressão Gênica/efeitos dos fármacos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Metformina/toxicidade , Fator de Transcrição STAT3/metabolismo , Alcaloides de Solanáceas/toxicidade , Fator de Transcrição Sp1/metabolismo , Células A549 , Animais , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Proteína Forkhead Box O3/antagonistas & inibidores , Proteína Forkhead Box O3/genética , Humanos , Imuno-Histoquímica , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Neoplasias Pulmonares , Metformina/uso terapêutico , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Alcaloides de Solanáceas/uso terapêutico , Transplante Heterólogo
2.
Cell Physiol Biochem ; 41(1): 339-357, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28214826

RESUMO

BACKGROUND: Emodin has anti-neoplastic activities on multiple tumors. However, the molecular mechanisms underlying this effect still remain to be fully understood. METHODS: Cell viability and cell cycle distribution were measured using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assays and flow cytometry, respectively. Cell invasion and migration were examined by transwell invasion and wound healing assays. Western blot analysis was performed to examine the phosphorylation and protein expression of AMP-activated protein kinase alpha (AMPKα), extracellular signaling-regulated kinase 1/2 (ERK1/2), peroxisome proliferators-activated receptor gamma (PPARγ), insulin-like growth factor (IGF) binding protein 1 (IGFBP1) and the transcription factor Sp1. QRT-PCR was used to examine the mRNA levels of the IGFBP1 gene. Small interfering RNAs (siRNAs) were used to knockdown PPARγ and IGFBP1 genes. Exogenously expression of IGFBP1 and Sp1 was determined by transient transfection assays. IGFBP1 promoter activity was measured by Secrete-Pair Dual Luminescence Assay Kit. In vivo nude mice xenograft model and bioluminescent imaging system were used to confirm the findings. RESULTS: We showed that emodin induced cell cycle arrest of NSCLC cells. Emodin increased PPARγ protein and luciferase reporter activity, which were abolished by inhibitors of MAPK extracellular signaling-regulated kinase (ERK) kinase (MEK)/ERK and AMPK. Silencing of PPARγ abrogated emodin-inhibited cell growth and cell cycle arrest. Furthermore, emodin elevated IGFBP1 mRNA, protein, and promoter activity through activation of PPARγ. Intriguingly, overexpressed Sp1 attenuated emodin-induced IGFBP1 expression, which was not observed in cells with silenced PPARγ gene. Moreover, silencing of IGFBP1 gene blunted emodin-induced inhibition of cell growth and cell cycle arrest. On the contrary, overexpressed IGFBP1 enhanced emodin-induced phosphorylation of AMPKα and ERK1/2, and restored emodin-inhibited growth in cells with silenced endogenous IGFBP1 gene. Emodin also inhibited growth of lung xenograft tumors and Sp1, and increased IGFBP1 and PPARγ protein expressions In vivo. CONCLUSION: Collectively, our results show that emodin inhibits growth of non-small-cell lung cancer (NSCLC) cells through ERK and AMPKα-mediated induction of PPARγ, followed by reduction of Sp1. This in turn induces IGFBP1 gene expression. Thus, the signaling cascades, positive feedback loop and cooperative interplay between transcription factors-induced the expression of IGFBP1 gene contribute to the overall responses of emodin. This study provides a novel mechanism by which emodin inhibits growth of human lung cancer cells.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Emodina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , PPAR gama/metabolismo , Fator de Transcrição Sp1/metabolismo , Células A549 , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Emodina/uso terapêutico , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , PPAR gama/antagonistas & inibidores , PPAR gama/genética , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo
3.
Neuro Oncol ; 17(8): 1076-85, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25543125

RESUMO

BACKGROUND: Signaling by insulin-like growth factor 1 receptor (IGF-1R) can contribute to the formation and progression of many diverse tumor types, including glioblastoma. We investigated the effect of the IGF-1R blocking antibody IMC-A12 on glioblastoma growth in different in vivo models. METHODS: U87 cells were chosen to establish rapidly growing, angiogenesis-dependent tumors in the brains of nude mice, and the GS-12 cell line was used to generate highly invasive tumors. IMC-A12 was administered using convection-enhanced local delivery. Tumor parameters were quantified histologically, and the functional relevance of IGF-1R activation was analyzed in vitro. RESULTS: IMC-A12 treatment inhibited the growth of U87 and GS-12 tumors by 75% and 50%, respectively. In GS-12 tumors, the invasive tumor extension and proliferation rate were significantly reduced by IMC-A12 treatment, while apoptosis was increased. In IMC-A12-treated U87 tumors, intratumoral vascularization was markedly decreased, and tumor cell proliferation was moderately reduced. Flow cytometry showed that <2% of U87 cells but >85% of GS-12 cells expressed IGF-1R. Activation of IGF-1R by IGF-1 and IGF-2 in GS-12 cells was blocked by IMC-A12. Both ligands stimulated GS-12 cell proliferation, and IGF-2 also stimulated migration. IMC-A12 inhibited these stimulatory effects and increased apoptosis. In U87 cells, stimulation with either ligand had no functional effect. CONCLUSIONS: IGF-1R blockade can inhibit glioblastoma growth by different mechanisms, including direct effects on the tumor cells as well as indirect anti-angiogenic effects. Hence, blocking IGF-1R may be useful to target both the highly proliferative, angiogenesis-dependent glioblastoma core component as well as the infiltrative periphery.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/fisiopatologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Glioblastoma/fisiopatologia , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/imunologia , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/fisiologia , Camundongos , Neovascularização Patológica , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Head Neck ; 35(1): 86-93, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22495823

RESUMO

BACKGROUND: Combined inhibition of epidermal growth factor receptor (EGFR) and insulin-like growth factor-1 receptor (IGF-1R) has been proposed as a therapy for cutaneous squamous cell carcinoma of the head and neck (SCCHN). METHODS: Receptor expression and downstream signaling were assessed in cutaneous squamous cell carcinoma (SCC) cell lines and patient samples. EGFR and IGF-1R signaling was inhibited in cutaneous SCC cell lines using erlotinib and/or picropodophyllin. RESULTS: EGFR and IGF-1R were overexpressed in cutaneous SCCHN specimens relative to normal skin. Dual inhibition of both receptors prevented cell growth and decreased activation of Akt and p42/44 mitogen-activated protein kinase (MAPK) more effectively than either inhibitor alone. CONCLUSION: Dual inhibition of EGFR and IGF-1R is effective at blocking cell growth, and is correlated with inhibition of Akt and p42/44 MAPK, suggesting that this may be a promising treatment for cutaneous SCCHN.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Cloridrato de Erlotinib , Imunofluorescência , Humanos , Podofilotoxina/análogos & derivados , Podofilotoxina/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Quinazolinas/uso terapêutico , Transdução de Sinais , Carcinoma de Células Escamosas de Cabeça e Pescoço
5.
Clin Obstet Gynecol ; 55(1): 156-72, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22343235

RESUMO

Ovarian cancer is the most lethal gynecologic cancer. Traditional therapies have included surgical management and cytotoxic chemotherapy; however, treatment paradigms continue to shift from empiric cytotoxic chemotherapy to more individualized treatment. Recent research efforts have focused on determining and targeting the molecular biological mechanisms of ovarian cancer in an attempt to develop novel therapeutic modalities with the ultimate goal of improving outcome while limiting toxicity. This chapter reviews progress in the development of novel therapies directed at major pathways implicated in ovarian tumorigenesis including angiogenesis, PARP inhibition, signal transduction, antifolate therapies, death receptor-mediated therapies, histone deacetylase inhibition, immunotherapeutics, and oncolytics.


Assuntos
Neoplasias Ovarianas/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais Murinos , Antineoplásicos/uso terapêutico , Bevacizumab , Antígeno Ca-125 , Ensaios Clínicos como Assunto , Docetaxel , Inibidores Enzimáticos/uso terapêutico , Feminino , Receptor 1 de Folato/antagonistas & inibidores , Genes BRCA1 , Genes BRCA2 , Terapia Genética , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Interleucina-12/uso terapêutico , Mutação , Proteína Oncogênica v-akt/antagonistas & inibidores , Neoplasias Ovarianas/irrigação sanguínea , Inibidores de Poli(ADP-Ribose) Polimerases , Inibidores de Proteínas Quinases/uso terapêutico , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Sirolimo/análogos & derivados , Sirolimo/uso terapêutico , Taxoides/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
6.
Curr Drug Targets ; 10(12): 1235-44, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19909234

RESUMO

Malignant mesothelioma is an aggressive tumour of the serosal surfaces with poor prognosis and increasing incidence due to widespread previous asbestos exposure. Relative chemotherapeutic and radiotherapeutic resistance makes malignant pleural mesothelioma (MPM) difficult to manage, even though encouraging results were achieved with multimodality treatment. Better knowledge of angiogenesis and molecular pathways involved in MPM seems to be the right way to define new targets for systemic treatment. Neoangiogenesis may be considered as a critical step in the development of mesothelioma. Vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF) are autocrine growth factors in MPM and epidermal growth factor receptor (EGFR) appears highly expressed in this tumour. Tyrosine kinase inhibitors (TKIs) targeting growth factors like vandetanib, dasatinib, and angiogenesis inhibitors like bevacizumab, are among the most promising agents under evaluation in clinical trials. Mesothelioma is a malignancy which owes its chemoresistance to an apoptotic defect. Thus the introduction of new biologic drugs like vorinostat, bortezomib, everolimus and temsirolimus, in the treatment of MPM finds a strong rationale. This review focuses on the current target therapies and evaluates future biologic approaches for the systemic management of MPM.


Assuntos
Antineoplásicos/uso terapêutico , Mesotelioma/tratamento farmacológico , Neoplasias Pleurais/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Animais , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/fisiologia , Fator de Crescimento de Hepatócito/fisiologia , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-met/fisiologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
8.
Reprod Biol Endocrinol ; 6: 6, 2008 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-18234122

RESUMO

BACKGROUND: The purpose of this study was to examine the effect of insulin on expression and synthesis of IGFBP-1 and IGFBP-2 in the baboon endometrium in vitro. METHODS: Baboon endometrial explants collected from cycling, ovariectomized, steroid-treated, simulated-pregnant and pregnant animals were cultured for 48 h in the presence or absence of insulin, with or without estradiol, progesterone and hCG. RESULTS: Insulin clearly inhibited IGFBP-1 production and mRNA expression in a time- and dose-dependent manner, whereas IGFBP-2 synthesis was not significantly affected. The inhibitory effects of insulin on IGFBP-1 were more evident in explants of non-pregnant tissue or tissue away from the implantation site. In the absence of insulin, synthesis of IGFBP-1 was induced in explants with low levels of de novo synthesis whereas IGFBP-2 synthesis was inhibited. This effect was potentiated by steroids and hCG in the explant cultures. CONCLUSION: Insulin differentially regulates endometrial IGFBP-1 and IGFBP-2 secretion in the baboon.


Assuntos
Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Insulina/farmacologia , Animais , Gonadotropina Coriônica/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Estradiol/farmacologia , Feminino , Técnicas In Vitro , Insulina/administração & dosagem , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Ciclo Menstrual , Ovariectomia , Papio , Gravidez , Progesterona/farmacologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , Esteroides/farmacologia
9.
Am J Physiol Endocrinol Metab ; 291(4): E835-42, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16720626

RESUMO

During perinatal development, the regulation of IGF system appears to be growth hormone (GH) independent. By using highly purified primary fetal hepatocytes, we investigated the role of prolactin (PRL) in the regulation of IGF system and hepatocyte proliferation. We also analyzed the consequence of a maternal low-protein (LP) diet on the regulation of IGF, IGF-binding protein (IGFBP), and hepatocyte proliferation by prolactin. Pregnant Wistar rats were fed a control (C) diet (20% protein) or isocaloric (LP; 8%) diet throughout gestation. On day 21.5, fetal hepatocytes were cultured for 4 days and incubated with rat prolactin. In the C hepatocytes, PRL at 100 ng/ml decreased the abundance of IGFBP-1 and IGFBP-2 by 50 (P < 0.05) and 60% (P < 0.01), respectively. It also reduced by 70% the level of IGF-II mRNA (P < 0.01). By contrast, PRL failed to modulate IGFBP-1 and IGFBP-2 production by LP hepatocytes, and this was associated with reduced abundance of the short form of PRL receptor (P < 0.05). PRL had no effect on either the proliferation or the IGF-I production by C and LP hepatocytes, although it reduced the expression of IGF-II. These results suggest that prolactin influences hepatocyte proliferation in vitro by inhibiting IGFBP-1, IGFBP-2, and IGF-II levels, which may coincide with the decline of IGF-II observed in rodents during late gestation in vivo. On the other hand, maternal LP diet induces a resistance of fetal hepatocytes to PRL.


Assuntos
Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Fígado/metabolismo , Desnutrição/metabolismo , Prolactina/farmacologia , Animais , Western Blotting , Processos de Crescimento Celular/fisiologia , Meios de Cultivo Condicionados/metabolismo , Proteínas Alimentares/metabolismo , Feminino , Desenvolvimento Fetal/fisiologia , Hepatócitos/citologia , Hepatócitos/metabolismo , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Fator de Crescimento Insulin-Like II/antagonistas & inibidores , Fator de Crescimento Insulin-Like II/genética , Fígado/citologia , Fígado/embriologia , Masculino , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Distribuição Aleatória , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Hum Reprod ; 19(7): 1513-8, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15155604

RESUMO

BACKGROUND: The purpose of this study was to determine the effect of progestin, relaxin (RLX) and transforming growth factor beta1 (TGFbeta1) on the content of relaxin receptor (LGR7) mRNA. The effect of RLX on insulin-like growth factor binding protein-1 (IGFBP-1) production was determined to evaluate the biological function of RLX/receptor in human endometrial cells. METHODS AND RESULTS: The levels of LGR7 mRNA and the effect of hormones were determined by real-time PCR in endometrial cells. LGR7 mRNA was found to be relatively abundant in endometrial glands and decidual cells and much less in endometrial stromal cells. In stromal cells, medroxyprogesterone acetate (MPA), or MPA plus RLX, significantly increased the LGR7 mRNA and RLX alone had little effect. In decidual cells, RLX increased LGR7 mRNA in a dose- and time-dependent fashion. TGFbeta1 reduced the LGR7 mRNA. In stromal cells, MPA alone caused a slight increase (2-4-fold) of the production rate of IGFBP-1 whereas MPA plus RLX synergistically increased (>40-fold) the IGFBP-1 production. In decidual cells in which the basal production rate was already approximately 50-fold higher than in stromal cells, RLX alone caused an additional increase (>30-fold) on the production rate. TGFbeta1 inhibited the IGFBP-1 production. CONCLUSION: The present study showed that in undifferentiated endometrial stromal cells, progestin increases the RLX receptor content to enhance the effect of RLX on the target gene (IGFBP-1). In decidual cells, RLX alone up-regulates its receptor, resulting in a large scale induction of IGFBP-1. TGFbeta1 has an inhibitory effect on LGR7 and IGFBP-1.


Assuntos
Endométrio/metabolismo , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Receptores de Peptídeos/antagonistas & inibidores , Receptores de Peptídeos/metabolismo , Relaxina/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Células Cultivadas , Decídua/citologia , Decídua/metabolismo , Sinergismo Farmacológico , Endométrio/citologia , Feminino , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Acetato de Medroxiprogesterona/antagonistas & inibidores , Acetato de Medroxiprogesterona/farmacologia , Proteínas de Membrana/genética , Progestinas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Relaxina/antagonistas & inibidores , Células Estromais/metabolismo , Fator de Crescimento Transformador beta1
11.
J Clin Endocrinol Metab ; 86(6): 2484-93, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11397844

RESUMO

A highly migratory subpopulation of the human placental trophoblast, known as the extravillous trophoblast (EVT), invades the uterus and its vasculature, to establish adequate exchange of key molecules between the maternal and fetal circulations. During their formation, EVT cells selectively acquire alpha 5 beta 1 integrin. We had shown that alpha 5 beta 1 is required for their migratory function, and that EVT cell migration is stimulated by insulin-like growth factor-binding protein (IGFBP)-1 produced by the uterine decidua. The present study examined whether this stimulation is dependent on binding of the Arg-Gly-Asp (RGD) domain of IGFBP-1 to an RGD binding site on the alpha 5 beta 1 integrin, followed by activation of focal adhesion kinase (FAK) and stimulation of the mitogen-activated protein kinase (MAPK) pathway. IGFBP-1 treatment increased migration of EVT cells, whereas an anti-alpha 5 beta 1 integrin antibody blocked migration regardless of IGFBP-1 treatment. Migration stimulation by IGFBP-1 was abrogated by pretreatment with a Gly-Arg-Gly-Asp-Ser-Pro (GRGDSP), but not a Gly-Arg-Gly-Glu-Ser-Pro (GRGESP) hexapeptide, and by mutation of the RGD domain of IGFBP-1 to Trp-Gly-Asp (WGD). IGFBP-1 treatment caused a rapid localization of immunoreactive FAK to cellular lamellipodia, a rapid increase in phosphorylation of FAK and extracellular-signal regulated kinases 1 and 2. Preincubation of EVT cells with Herbimycin A, a tyrosine kinase inhibitor, abrogated IGFBP-1 effects; whereas an MAPK kinase inhibitor, PD 98059, reduced migration regardless of IGFBP-1 treatment. These results indicate that IGFBP-1 stimulation of EVT cell migration occurs by binding of its RGD domain to the alpha 5 beta 1 integrin, leading to activation of FAK and stimulation of MAPK pathway.


Assuntos
Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/farmacologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Receptores de Fibronectina/fisiologia , Transdução de Sinais/fisiologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/fisiologia , Animais , Anticorpos/farmacologia , Benzoquinonas , Células CHO , Divisão Celular/fisiologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Cricetinae , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Imunofluorescência , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Lactamas Macrocíclicas , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oligopeptídeos/metabolismo , Fosforilação , Estrutura Terciária de Proteína/fisiologia , Proteínas Tirosina Quinases/metabolismo , Quinonas/farmacologia , Receptores de Fibronectina/genética , Receptores de Fibronectina/imunologia , Rifabutina/análogos & derivados , Fatores de Tempo , Distribuição Tecidual , Trofoblastos/citologia
12.
J Clin Endocrinol Metab ; 86(5): 2060-4, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11344207

RESUMO

In human pregnancy, insulin-like growth factor (IGF)-II messenger RNA (mRNA) is expressed at the maternal-fetal interface exclusively by the placental trophoblast. Highest levels are expressed by the invading extravillous trophoblasts, which also secrete matrix metalloproteinases as they degrade the decidual extracellular matrix. In contrast, the maternal decidua expresses high levels of IGF-binding protein (IGFBP)-1 and tissue inhibitors of matrix metalloproteinase (TIMPs), both of which inhibit trophoblast invasiveness in vitro. The present study investigated the hypothesis that IGF-II may serve as a paracrine modulator of maternal restraints on invasion, by examining its effects on TIMP-3 and IGFBP-1 expression by decidualized endometrial stromal cells. Human endometrial stromal cells were decidualized in vitro with progesterone (P), after which 0-130 nM IGF-II and IGF analogs were added. IGFBP-1 in conditioned medium was assayed by immunoradiometric assay. In addition, Northern analyses were conducted using a PCR-generated 421-bp complementary DNA (cDNA) fragment corresponding to nucleotides 132-553 of the human TIMP-3 cDNA, and a 934-bp EcoRI fragment of the human IGFBP-1 cDNA. TIMP-3 mRNA transcripts of 2.2, 2.5, and 4.4 kilobases were detected in decidualized stromal cells not treated with IGF-II, but not detected in nondecidualized stromal cells, consistent with its known induction upon decidualization and in response to P. In decidualized stromal cells, IGF-II and Des(1-6) IGF-II, an analog with reduced affinity for IGFBPs, caused a dose-dependent inhibition of TIMP-3 mRNA expression. Long R(3) IGF-I, an IGF analog with minimal affinity for IGFBPs, also significantly inhibited (79 +/- 0.3%) TIMP-3 mRNA expression in these cells at 6 nM. Decidualized stromal cells secreted IGFBP-1 and expressed a 1.5-kilobase IGFBP-1 transcript, which was not detected in nondecidualized cells, consistent with its known induction upon decidualization and in response to P. IGF-II caused a dose-dependent inhibition of IGFBP-1 mRNA expression and protein secretion in decidualized stromal cells when added in molar excess of endogenous IGFBP-1 levels, with virtually complete inhibition at higher concentrations of IGF-II (65 and 130 nM). By comparison, Long R(3) IGF-I inhibited IGFBP-1 expression with a 50% effective dose of 0.2-0.4 nM. These data suggest that the invading trophoblast has the capacity, via IGF-II, to inhibit maternal restraints on trophoblast invasiveness by regulating decidual TIMP-3 and IGFBP-1.


Assuntos
Decídua/fisiologia , Implantação do Embrião , Endométrio/efeitos dos fármacos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Fator de Crescimento Insulin-Like II/farmacologia , Inibidor Tecidual de Metaloproteinase-3/antagonistas & inibidores , Trofoblastos/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , RNA Mensageiro/análise , Células Estromais/efeitos dos fármacos , Inibidor Tecidual de Metaloproteinase-3/genética
13.
J Biol Chem ; 275(10): 7289-95, 2000 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-10702299

RESUMO

The forkhead rhabdomyosarcoma transcription factor (FKHR) is a promising candidate to be the transcription factor that binds to the insulin response element of the insulin-like growth factor-binding protein-1 (IGFBP-1) promoter and mediates insulin inhibition of IGFBP-1 promoter activity. Cotransfection of mouse FKHR increased IGFBP-1 promoter activity 2-3-fold in H4IIE rat hepatoma cells; insulin inhibited FKHR-stimulated promoter activity approximately 70%. A C-terminal fragment of mouse FKHR (residues 208-652) that contains the transcription activation domain fused to a Gal4 DNA binding domain potently stimulated Gal4 promoter activity. Insulin inhibited FKHR fragment-stimulated promoter activity by approximately 70%. Inhibition was abolished by coincubation with the phosphatidylinositol-3 kinase inhibitor, LY294002. The FKHR 208-652 fragment contains two consensus sites for phosphorylation by protein kinase B (PKB)/Akt, Ser-253 and Ser-316. Neither site is required for insulin inhibition of promoter activity stimulated by the FKHR fragment, and overexpression of Akt does not inhibit FKHR fragment-stimulated Gal4 promoter activity. These results suggest that insulin- and phosphatidylinositol-3 kinase-dependent phosphorylation of another site in the fragment by a kinase different from PKB/Akt inhibits transcription activation by the fragment. Phosphorylation of this site also may be involved in insulin inhibition of transcription activation by full-length FKHR, but only after phosphorylation of Ser-253 by PKB/Akt.


Assuntos
Proteínas de Ligação a DNA/farmacologia , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Insulina/farmacologia , Proteínas do Tecido Nervoso , Fragmentos de Peptídeos/farmacologia , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases , Fatores de Transcrição/farmacologia , Ativação Transcricional/efeitos dos fármacos , Animais , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Camundongos , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-akt , RNA Mensageiro/análise , Ratos
14.
Endocrinology ; 140(9): 3962-70, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10465265

RESUMO

Insulin-like growth factor-binding protein-1 (BP-1) is a multifunctional protein that binds IGF-I in solution and integrins on the cell surface. BP-1 is overexpressed during catabolic illnesses, and the protein accumulates in skeletal muscle. To define a potential physiological role for BP-1 in regulating muscle protein balance, we have examined the effect of IGF-I and BP-1 on protein synthesis and degradation in human skeletal muscle cells. IGF-I-stimulated protein synthesis by 20%, and this was completely inhibited by either phosphorylated or nonphosphorylated BP-1. Half-maximal inhibition of protein synthesis occurred at a molar ratio of BP-1 to IGF-I of 1.5:1. BP-1 failed to form a complex with a truncated form of IGF-I (desIGF-I), and consequently, BP-1 failed to inhibit the ability of desIGF-I to stimulate protein synthesis. IGF-I and BP-1 dose-dependently inhibited protein degradation individually, and both BP-1 phosphovariants failed to block the ability of IGF-I to do the same. Blocking integrin receptor occupancy with the integrin antagonist echistatin blunted the ability of BP-1 to inhibit protein degradation, but had no significant effect on IGF-I-mediated changes in protein synthesis or degradation. The extracellular matrix protein vitronectin also inhibited protein degradation, but vitronectin receptor antibodies failed to block BP-1 action. In contrast, antibodies to the beta1 integrin subunit blocked BP-1-mediated inhibition of protein degradation. Rapamycin inhibited IGF-I-dependent protein synthesis, but not the ability of IGF-I to inhibit proteolysis. In contrast, rapamycin completely blocked the ability of BP-1 to inhibit proteolysis. Our results demonstrate that BP-1 inhibits IGF-I-mediated protein synthesis by binding to IGF-I. BP-1, acting independently of IGF-I, inhibits protein degradation. The IGF-independent response occurs via beta1 integrin binding and stimulation of a rapamycin-sensitive signal transduction pathway.


Assuntos
Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Androstadienos/farmacologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Glucose/metabolismo , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/metabolismo , Integrina beta1/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/biossíntese , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Peptídeos/farmacologia , Receptores de Somatomedina/fisiologia , Sirolimo/farmacologia , Wortmanina
15.
Biochemistry ; 37(25): 8870-8, 1998 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-9636028

RESUMO

IGF-1 (insulin-like growth factor 1) is a 70-residue protein hormone which has both metabolic and mitogenic activities mediated through IGF-1 binding to cell surface receptors. However, an unrelated class of proteins, the IGF-binding proteins (IGFBPs) also bind IGF-1 in the serum and tissues and block or modulate its activity in vivo. Therefore, inhibitors of the IGFBPs can alter the distribution between free and bound IGF-1 [Loddick, S. A., Liu, X.-J., Lu, Z.-X., Liu, C., Behan, D. P., Chalmers, D. C., Foster, A. C., Vale, W. W., Ling, N., and De Souza, E. B. (1998) Proc. Natl. Acad. Sci. U.S.A. 95, 1894-1898] and potentially affect the distribution of IGF-1 among body tissues. We report here that phage-displayed peptide libraries have yielded a peptide that binds IGFBP-1 and produces IGF-like activity at sub-micromolar concentrations. The 14-residue peptide has an extremely well-defined solution conformation that can aid in the design of smaller, orally active compounds. Interestingly, the peptide structure contains a helix, as does one region of IGF-1 previously implicated in IGFBP binding, yet displays side chains different from those of the IGF-1 helix I. Furthermore, an IGF-1 variant lacking receptor-signaling activity in vitro is shown here to produce IGF-like mitogenic and metabolic activity in vivo. These results suggest that small antagonist mimetics of protein ligands, identified by binding selection to otherwise inhibitory factors, may be useful as indirect agonists for a variety of therapeutic applications.


Assuntos
Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/metabolismo , Mimetismo Molecular , Sequência de Aminoácidos , Animais , Bacteriófago M13/metabolismo , Ligação Competitiva/efeitos dos fármacos , Insulina/farmacologia , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/química , Fator de Crescimento Insulin-Like I/farmacologia , Ligantes , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Biblioteca de Peptídeos , Peptídeos/síntese química , Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ratos Zucker , Receptor IGF Tipo 1/deficiência
16.
J Clin Invest ; 100(10): 2596-605, 1997 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9366575

RESUMO

IGFs are pleiotrophic mitogens for porcine smooth muscle cells (pSMC) in culture. The effects of IGFs on cells are modulated by various insulin-like growth factor-binding proteins (IGFBP). IGFBP-5 is synthesized by pSMC and binds to the extracellular matrix. However, IGFBP-5 is also secreted into conditioned medium of cultured cells and is cleaved into fragments by a concomitantly produced protease. These fragments have reduced affinity for the IGFs and cleavage makes it difficult to assess the role of intact IGFBP-5. To study the consequence of accumulation of intact IGFBP-5 in medium, we determined the cleavage site in IGFBP-5 and prepared a protease resistant mutant. Amino acid sequencing of purified IGFBP-5 fragments suggested Arg138-Arg139 as the primary cleavage site. Arg138-Arg139-->Asn138-Asn139 mutations were introduced to create protease-resistant IGFBP-5, which has the same affinity for IGF-I as the native protein. This mutant IGFBP-5 remained intact even after 24 h of incubation and it inhibited several IGF-I actions when added to pSMC culture medium. The mutant IGFBP-5 (500 ng/ml) decreased IGF-I stimulated cellular DNA synthesis by 84%, protein synthesis by 77%, and it inhibited IGF-I stimulated migration of pSMC by 77%. It also inhibited IGF-I stimulation of IRS-1 phosphorylation. In contrast, the same amount of native IGFBP-5 did not inhibit IGF-I actions. The significance of inhibitory effects of the protease resistant IGFBP-5 was further demonstrated in pSMC transfected with mutant or native IGFBP-5 cDNAs. The mutant IGFBP-5 accumulated in culture medium of transfected cells, while native IGFBP-5 was degraded into fragments, PSMC overexpressing the mutant IGFBP-5 also responded poorly to IGF-I compared with mock transfected cells. IGF-I (5 ng/ml) increased [35S]methionine incorporation into control cells by 36% above the basal level, but it did not significantly change (4%) in pSMC cultures that were producing the mutant IGFBP-5. In conclusion, the accumulation of protease-resistant IGFBP-5 in the medium was inhibitory to IGF-I actions on pSMC. This suggests that proteolysis can prevent IGFBP-5 from acting as an inhibitor of IGF-I-stimulated effects and that it serves as an important mechanism for regulating cellular responsiveness to IGF-I.


Assuntos
Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/farmacologia , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Metaloendopeptidases/metabolismo , Músculo Liso Vascular/fisiologia , Sequência de Aminoácidos , Animais , Aorta , Arginina , Asparagina , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/química , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Mutação Puntual , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Suínos , Timidina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...