Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Int J Oncol ; 31(2): 441-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17611702

RESUMO

Lysophosphatidic acid (LPA) is an important intercellular signaling molecule involved in a myriad of biological responses. Elevated concentrations of LPA are present in the ascites and plasma of ovarian cancer patients suggesting a role for LPA in the pathophysiology of ovarian cancer. We have demonstrated previously that oleoyl (18:1) LPA at concentrations present in ascites induces the secretion of urokinase plasminogen activator (uPA) from ovarian cancer cells, possibly linking LPA to cellular invasion. In this study we sought to elucidate which signaling pathway(s) are involved in LPA-mediated secretion of uPA from ovarian cancer cells. Specific inhibitors were utilized to determine if interference with the p38(MAPK), p42/44(MAPK), and PI3K pathways functionally blocked LPA-mediated uPA secretion. LPA stimulation of ovarian cancer cells markedly increased the phosphorylation and activity of p38(MAPK), p42/p44(MAPK), and PI3K. Both tyrosine phosphorylation and Src kinase activity were required for optimal activation of signaling by LPA including phosphorylation of p38(MAPK). Inhibition of p38(MAPK) signaling by SB202190 completely abrogated LPA-induced uPA secretion, while inhibition of the p42/44(MAPK) or PI3K pathways with PD98059 or wortmannin and LY294002, respectively, decreased but did not completely block uPA secretion. In contrast, inhibitors of phospholipase D or the p70S6 kinase pathway did not alter LPA-induced uPA secretion. Further, tyrosine phosphorylation and functional Src were required for optimal uPA secretion. Finally, LPA induces uPA secretion from ovarian cancer cells predominantly through the LPA2 receptor, with LPA3 contributing to this process. These results indicate that the p38(MAPK) signaling pathway is required for optimal LPA-dependent uPA secretion from ovarian cancer cells.


Assuntos
Lisofosfolipídeos/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Transporte Biológico , Diferenciação Celular , Linhagem Celular Tumoral , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Sistema de Sinalização das MAP Quinases , Neoplasias Ovarianas/metabolismo , Fosfolipase D/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais
2.
Biochim Biophys Acta ; 1582(1-3): 257-64, 2002 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-12069836

RESUMO

Lysophosphatidic acid (LPA) is a naturally occurring phospholipid that exhibits pleiotrophic biological activities, ranging from rapid morphological changes to long-term cellular effects such as induction of gene expression and stimulation of cell proliferation and survival on a wide spectrum of cell types. LPA binds and activates distinct members of the Edg/LP subfamily of G protein-coupled receptors that link to multiple G proteins including Gi, Gq and G12/13 to elicit cellular responses. LPA plays a critical role as a general growth, survival and pro-angiogenic factor, in the regulation of physiological and pathophysiological processes in vivo and in vitro. Our previous work indicates that abnormalities in LPA metabolism and function in ovarian cancer patients may contribute to the initiation and progression of the disease. Thus, LPA could be a potential target for cancer therapy. This review summarizes evidence that implicates LPA in the pathophysiology of human ovarian cancer and likely other types of human malignancies.


Assuntos
Lisofosfolipídeos/fisiologia , Neoplasias Ovarianas/fisiopatologia , Receptores Acoplados a Proteínas G , Feminino , Humanos , Receptores de Superfície Celular/fisiologia , Receptores de Ácidos Lisofosfatídicos , Valores de Referência
3.
Semin Oncol ; 30(5 Suppl 16): 93-104, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14613030

RESUMO

Modulation of the signaling pathways that are aberrant in cancer cells has the potential to provide an effective nontoxic approach to patient management in a broad range of cancers. This quest has taken a major leap forward with the demonstration that STI-571 (imatinib mesylate) induces clinical and molecular remissions in the majority of patients with interferon-refractory chronic myelogenous leukemia and gastrointestinal stromal tumors through inhibition of the Bcr/Abl fusion protein required for the initiation and progression of chronic myelogenous leukemia and inhibition of a mutant, activated c-kit present in gastrointestinal stromal tumors. Support for the concept of targeting products of fusion genes found in specific cancers was first provided by the efficacy of all-trans retinoic acid in acute promyelocytic leukemia where the RARalpha all-trans retinoic acid target is the target of multiple different chromosomal rearrangements. In breast cancer, trastuzumab, which alters the function of the HER2 proto-oncogene overexpressed in a portion of breast cancers, provides an additional example of targeting specific molecular aberrations present in cancer cells. Although the target for these signal transduction modulators is functional in normal cells, acceptable therapeutic indices sufficient to prevent tumor growth without unacceptable toxicities have been observed. Whether STI-571 and other signal transduction modulators also target the stroma, and specifically the neovasculature, in addition to the tumor remains an open question. The presence of the target in the cancer cells or in the surrounding stroma appears to be required but not sufficient for the action of molecular therapeutics. Thus, linking molecular diagnostics to identify patients where the target is amplified or activated and driving the pathophysiology of the patients' tumor to effective molecular therapeutics will be necessary to translate these concepts into approaches that will alter the outcome for breast cancer patients. This review will focus on the phosphatidylinositol 3-kinase pathway and novel molecules targeting this pathway to illustrate the questions and challenges underlying the implementation of molecular therapeutics in breast cancer.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases/antagonistas & inibidores , Animais , Benzamidas , Neoplasias da Mama/enzimologia , Cromonas/uso terapêutico , Ensaios Clínicos como Assunto , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Mesilato de Imatinib , Morfolinas/uso terapêutico , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases , Piperazinas/uso terapêutico , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas , Proteínas Proto-Oncogênicas c-akt , Pirimidinas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Proteínas Supressoras de Tumor
4.
Cancer Treat Res ; 107: 259-83, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11775454

RESUMO

Lysophosphatidic acid (LPA), the simplest of all phospholipids, exhibits pleiomorphic functions in multiple cell lineages. The effects of LPA appear to be mediated by binding of LPA to specific members of the endothelial differentiation gene (Edg) family of G protein-coupled receptors (GPCR). Edg 2, Edg4, and Edg7 are high affinity receptors for LPA, and Edg1 may be a low affinity receptor for LPA. PSP24 has been shown to be responsive to LPA in Xenopus oocytes, however, its role in mammalian cells is unclear. The specific biochemical events initiated by the different Edg receptors, as well as the biological outcomes of activation of the individual receptors, are only beginning to be determined. LPA levels are consistently elevated in the plasma and ascites of ovarian cancer patients, but not in most other epithelial tumors, with the exception of cervix and endometrium, suggesting that LPA may be of particular importance in the pathophysiology of ovarian cancer. In support of this concept, ovarian cancer cells constitutively and inducibly produce high levels of LPA and demonstrate markedly different responses to LPA than normal ovarian surface epithelium. Edg4 and Edg7 levels are consistently increased in malignant ovarian epithelial cells contributing to the aberrant response of ovarian cancer cells to LPA. Edg2 may represent a negative regulatory LPA receptor inducing apoptosis in ovarian cancer cells. Thus, increased levels of LPA, altered receptor expression and altered responses to LPA may contribute to the initiation, progression or outcome of ovarian cancer. Over 40% of known drugs target GPCR, making LPA receptors attractive targets for molecular therapeutics. Indeed, using the structure-function relationship of LPA in model systems, we have identified selective Edg2 anatgonists, as well as Edg4 and Edg7 agonists. These lead compounds are being assessed in preclinical model systems. Understanding the mechanisms regulating LPA production, metabolism and function could lead to improved methods for early detection and to new targets for therapy in ovarian cancer.


Assuntos
Biomarcadores Tumorais/análise , Transformação Celular Neoplásica , Lisofosfolipídeos/farmacologia , Neoplasias Ovarianas/fisiopatologia , Divisão Celular/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Lisofosfolipídeos/análise , Lisofosfolipídeos/biossíntese , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/tratamento farmacológico , Transdução de Sinais
5.
Proc Natl Acad Sci U S A ; 102(35): 12519-24, 2005 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-16116079

RESUMO

We show that atypical PKCiota, which plays a critical role in the establishment and maintenance of epithelial cell polarity, is genomically amplified and overexpressed in serous epithelial ovarian cancers. Furthermore, PKCiota protein is markedly increased or mislocalized in all serous ovarian cancers. An increased PKCiota DNA copy number is associated with decreased progression-free survival in serous epithelial ovarian cancers. In a Drosophila in vivo epithelial tissue model, overexpression of persistently active atypical PKC results in defects in apical-basal polarity, increased Cyclin E protein expression, and increased proliferation. Similar to the Drosophila model, increased PKCiota proteins levels are associated with increased Cyclin E protein expression and proliferation in ovarian cancers. In nonserous ovarian cancers, increased PKCiota protein levels, particularly in the presence of Cyclin E, are associated with markedly decreased overall survival. These results implicate PKCiota as a potential oncogene in ovarian cancer regulating epithelial cell polarity and proliferation and suggest that PKCiota is a novel target for therapy.


Assuntos
Ciclina E/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Animais , Animais Geneticamente Modificados , Apoptose , Polaridade Celular , Proliferação de Células , Drosophila/citologia , Drosophila/enzimologia , Drosophila/genética , Olho/citologia , Olho/enzimologia , Olho/crescimento & desenvolvimento , Feminino , Amplificação de Genes , Humanos , Oncogenes , Neoplasias Ovarianas/genética , Prognóstico , Ratos
6.
Gynecol Oncol ; 85(3): 451-8, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12051873

RESUMO

OBJECTIVE: One potential limitation of gene therapy for epithelial tumors is the lack of tissue or tumor specificity of treatment. Tumor-selective expression of gene therapies may avoid deleterious side effects and improve the efficacy of the treatment. The aim of this study was to evaluate the tissue and tumor specificity of four different potential gene therapy promoters, to determine their usefulness in tissue-specific gene therapy of epithelial ovarian carcinomas. METHODS: Three potential epithelial cell-selective (hESE1, SLP1, OSP1) and one potential tumor-selective (hTERT) promoter were placed upstream of a luciferase construct to determine relative activity in a wide variety of normal and malignant cell lines. Transient transfection and luciferase assays were carried out in 12 epithelial ovarian (3 SV40 T antigen-transfected normal and 9 malignant) and 8 control cell lines. RESULTS: Luciferase assays revealed that the hTERT promoter presented the highest tumor selectivity. hESE1 and SLP1 promoters showed strong epithelial cell selectivity (hESE1, 16/17; SLP1, 15/17), with the OSP1 (11/17) promoter exhibiting lower epithelial selectivity. Of the potential promoters for gene therapy, hTERT promoter exhibited the strongest transcriptional activity in most of the tumor cell lines. None of the promoters exhibited strict ovarian epithelium selectivity. CONCLUSION: The hTERT promoter may be an optimal promoter for a univector gene therapy approach based on its high tumor selectivity. Utilization of multiple epithelial cell-specific promoters may result in a more tissue-selective gene therapy approach. Using a combination of promoters may prevent potential problems due to expression in nonepithelial stem cells that may constitutively express hTERT.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular , Terapia Genética/métodos , Neoplasias Ovarianas/genética , Regiões Promotoras Genéticas/genética , Animais , Células COS , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Proteínas de Ligação a DNA , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/genética , Genes Reporter/genética , Humanos , Luciferases/biossíntese , Luciferases/genética , Luciferases/metabolismo , Neoplasias Ovarianas/terapia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Telomerase/genética , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas
7.
J Biol Chem ; 278(14): 11962-9, 2003 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-12554733

RESUMO

Lysophosphatidic acid (LPA) is a bioactive lysophospholipid mediator that acts through G protein-coupled receptors. Most cell lines in culture express one or more LPA receptors, making it difficult to assign a response to specific LPA receptors. Dissection of the signaling properties of LPA has been hampered by lack of LPA receptor subtype-specific agonists and antagonists. The present study characterizes an ester-linked thiophosphate derivative (1-oleoyl-2-O-methyl-rac-glycerophosphothionate, OMPT) of LPA. OMPT is a functional LPA analogue with potent mitogenic activity in fibroblasts. In contrast to LPA, OMPT does not couple to the pheromone response through the LPA(1) receptor in yeast cells. OMPT induces intracellular calcium increases efficiently in LPA(3) receptor-expressing Sf9 cells but poorly in LPA(2) receptor-expressing cells. Guanosine 5'-O-(3-[(35)S]thio)triphosphate binding assays in mammalian cells showed that LPA exhibits agonistic activity on all three LPA receptor subtypes, whereas OMPT has a potent agonistic effect only on the LPA(3) receptor. In transiently transfected HEK293 cells, OMPT stimulates mitogen-activated protein kinases through the LPA(3) but not the LPA(1) or LPA(2) receptors. Furthermore, OMPT-induced intracellular calcium mobilization in mammalian cells is efficiently inhibited by the LPA(1)/LPA(3) receptor-selective antagonist VPC12249. These results establish that OMPT is an LPA(3)-selective agonist. OMPT binding to the LPA(3) receptor in mammalian cells is sufficient to elicit multiple responses, including activation of G proteins, calcium mobilization, and activation of mitogen-activated protein kinases. Thus OMPT offers a powerful probe for the dissection of LPA signaling events in complex mammalian systems.


Assuntos
Lisofosfolipídeos/farmacologia , Organotiofosfatos/farmacologia , Ácidos Fosfatídicos/farmacologia , Receptores de Superfície Celular/agonistas , Receptores Acoplados a Proteínas G , Transdução de Sinais/efeitos dos fármacos , Células 3T3 , Animais , Cálcio/metabolismo , Humanos , Rim/citologia , Lisofosfolipídeos/química , Mamíferos , Camundongos , Camundongos Endogâmicos C3H , Mitógenos/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Ácidos Lisofosfatídicos , Spodoptera , Leveduras/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA