Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Br J Pharmacol ; 163(2): 220-33, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21232046

RESUMO

The endothelins (ET) are a group of proteins that act through G-protein coupled receptors. Endothelin-1 (ET-1) was initially identified as a potent vasoconstrictor and dysregulation of the ET axis contributes to pathological processes responsible for cardiovascular disease states. More recently, the ET axis, in particular ET-1 acting through the endothelin A receptor (ET(A) ), has been implicated in the development of several cancers through activation of pathways involved in cell proliferation, migration, invasion, epithelial-mesenchymal transition, osteogenesis and angiogenesis. The endothelin B receptor (ET(B) ) may counter tumour progression by promoting apoptosis and clearing ET-1; however, it has recently been implicated in the development of some tumour types including melanomas and oligodendrogliomas. Here, we review emerging preclinical and clinical data outlining the role of the ET axis in cancer, and its antagonism as an attractive and challenging approach to improve clinical cancer management. Clinical data of ET(A) antagonists in patients with prostate cancer are encouraging and provide promise for new ET(A) antagonist-based treatment strategies. Given the unexpected opportunities to affect pleiotrophic tumorigenic signals by targeting ET(A)-mediated pathways in a number of cancers, the evaluation of ET-targeted therapy in cancer warrants further investigation.


Assuntos
Antagonistas do Receptor de Endotelina A , Antagonistas do Receptor de Endotelina B , Endotelinas/fisiologia , Neoplasias/terapia , Animais , Endotelina-1/fisiologia , Endotelina-2/fisiologia , Endotelina-3/fisiologia , Humanos , Terapia de Alvo Molecular , Neoplasias/metabolismo , Neoplasias/patologia , Receptor de Endotelina A/fisiologia , Receptor de Endotelina B/fisiologia
2.
J Neurosci Res ; 64(6): 636-45, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11398188

RESUMO

Nerve growth factor (NGF) binds to the TrkA tyrosine kinase and the p75 neurotrophin receptors. Depending upon which receptor is activated, NGF can induce differentiation or apoptosis. C6-2B glioma cells express the p75 receptor, but NGF decreases their growth only when TrkA is introduced (C6trk). It is unclear, however, whether TrkA reduces C6-2B cell growth by apoptosis or differentiation. To examine which mechanisms account for the anti-proliferative effect of NGF in these cells, we first analyzed whether NGF causes apoptosis by flow cytometry, two-site immunoassay and in situ TUNEL. None of these methods indicated that C6trk undergo apoptosis. Additional apoptotic markers, such as Bcl-2, Bax, Bad, p53, caspase 3, and NF-kappaB were also used. C6trk cells exhibited lower levels of Bcl-2 compared with the parental C6 mock cells, but no changes in the levels of other apoptotic proteins. Moreover, NGF increased AP-1 binding activity in C6trk cells, suggesting that NGF may induce differentiation. We then examined whether TrkA changes the glioma phenotype. In C6trk cells, but not in C6mock cells, NGF enhanced the levels of neuron-specific enolase as well as the levels of A2B5 and 2', 3'-cyclic nucleotide 3'-phosphodiesterase, markers for oligodendrocytes, without affecting the expression of other neuronal markers. Our data suggest that the antiproliferative properties of TrkA may rely on its ability to induce differentiation of C6 cells from undifferentiated glioma to oligodendrocytes.


Assuntos
Glioma , Oligodendroglia/citologia , Receptor trkA/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Marcação In Situ das Extremidades Cortadas , NF-kappa B/análise , NF-kappa B/metabolismo , Fator de Crescimento Neural/farmacologia , Oligodendroglia/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/análise , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Receptor de Fator de Crescimento Neural/análise , Receptor de Fator de Crescimento Neural/metabolismo , Fator de Transcrição AP-1/metabolismo , Células Tumorais Cultivadas/química , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/metabolismo , Proteína Supressora de Tumor p53/análise , Proteína X Associada a bcl-2
3.
Prostate ; 47(2): 102-10, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11340632

RESUMO

BACKGROUND: Fatty acid synthase (FAS) performs the anabolic conversion of dietary carbohydrate or protein to fat. FAS expression is low in most normal tissues, but is elevated in many human cancers, including androgen-sensitive and androgen-independent prostate cancer. METHODS: Immunohistochemical evaluation of FAS expression was performed in human prostate cancer specimens under various states of androgen ablation. In vitro and in vivo prostate cancer models were evaluated for FAS expression and activity under androgenic and androgen-depleted conditions, and were tested for sensitivity to antimetabolite drugs that target fatty acid synthesis. RESULTS: While FAS expression in the prostate was androgen responsive, it persisted or was reactivated in human prostate carcinoma after androgen ablation, and was high in 82% of lethal tumors examined at autopsy. Similar patterns of FAS expression and fatty acid synthesis were seen in cell culture and xenograft models of human prostate cancer. Pharmacologic inhibition of FAS resulted in a dose-dependent reduction of tumor growth in these models, including fourfold inhibition of an androgen-independent human prostate cancer xenograft with little associated toxicity. CONCLUSIONS: The data suggest that FAS expression/FA synthesis provides an important functional aspect of the malignant phenotype in prostate cancer, perhaps supporting cell growth or survival. FAS expression may be upregulated by alternate signaling pathways important for prostate cancer growth under androgen withdrawal. The re-emergence of FAS expression and activity during the development of androgen independence demonstrate that FAS may serve as a novel target for antimetabolite therapy in prostate cancer.


Assuntos
4-Butirolactona/análogos & derivados , Androgênios/fisiologia , Ácido Graxo Sintases/biossíntese , Neoplasias da Próstata/enzimologia , 4-Butirolactona/farmacologia , Animais , Anticorpos Monoclonais , Antineoplásicos/farmacologia , Western Blotting , Cromatografia em Camada Fina , Inibidores Enzimáticos/farmacologia , Ácido Graxo Sintases/antagonistas & inibidores , Humanos , Imuno-Histoquímica , Lipídeos/análise , Masculino , Camundongos , Camundongos Nus , Neoplasias Hormônio-Dependentes/enzimologia , Distribuição Aleatória , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cancer Res ; 61(9): 3599-603, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11325827

RESUMO

In this study, a panel of normal human prostate cells (HPCs) and tumor cells derived from metastases were studied by (1)H NMR spectroscopy to determine whether the malignant transformation of HPCs results in the elevation of choline compounds. Although an elevated choline signal has been observed previously in clinical studies, the contribution of the different Cho compounds to this elevation, as well as their quantification, has not been established until now. Here we have shown that HPCs derived from metastases exhibit significantly higher phosphocholine as well as glycerophosphocholine levels compared with normal prostate epithelial and stromal cells. Thus the elevation of the choline peak observed clinically in prostate cancer is attributable to an alteration of phospholipid metabolism and not simply to increased cell density, doubling time, or other nonspecific effects. Androgen deprivation of the androgen receptor-positive cell lines resulted in a significant increase of choline compounds after chronic androgen deprivation of the LNCaP cell line and in a decrease of choline compounds after a more acute androgen deprivation of the LAPC-4 cell line. These data strongly support the use of proton magnetic resonance spectroscopic imaging to detect the presence of prostate cancer for diagnosis, to detect response subsequent to androgen ablation therapy, and to detect recurrence.


Assuntos
Transformação Celular Neoplásica/metabolismo , Colina/metabolismo , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Androgênios/deficiência , Androgênios/fisiologia , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Masculino , Metástase Neoplásica , Neoplasias Hormônio-Dependentes/metabolismo , Ressonância Magnética Nuclear Biomolecular , Fosfatidilcolinas/metabolismo , Fosforilcolina/metabolismo , Próstata/patologia , Neoplasias da Próstata/patologia , Receptores Androgênicos/metabolismo , Receptores Androgênicos/fisiologia
5.
Prostate ; 40(4): 269-73, 1999 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10420156

RESUMO

BACKGROUND: Increased expression of caveolin has been associated with prostate cancer progression. In a mouse reconstitution model of prostate cancer, expression of caveolin was inversely related to androgen sensitivity: androgen independent clones had high caveolin expression; low caveolin expression was associated with sensitivity to androgen withdrawal. In contrast, several independent observations support the hypothesis that caveolin functions as a tumor suppressor. METHODS: Caveolin expression was studied by Western blot analysis and/or immunohistochemistry in three androgen-sensitive human prostate cancer cell lines LNCaP, LAPC-4 and LAPC-9, and following acute and chronic androgen deprivation, and in benign prostate epithelial cells. Expression of caveolin, androgen receptor (AR) and prostate specific antigen (PSA) was also examined after reintroduction of androgen. RESULTS: LNCaP grown continuously under androgen-depleted conditions for 6 to 42 months produced several androgen-independent and tumorigenic clones: tumors formed in 13/15 castrate and 7/15 intact male athymic nu/nu mice, but no tumors formed in wildtype LNCaP-bearing animals. Caveolin expression was decreased in every androgen-deprived LNCaP clone and following 150 days of androgen deprivation in LAPC-4. Caveolin expression by LAPC-9 was very low. Following exposure to dihydrotestosterone in vitro, caveolin and PSA expression increased within three days in the androgen-deprived clones of LNCaP. Benign prostate epithelial cells have high caveolin expression. CONCLUSIONS: Unlike the mouse reconstitution model of prostate cancer, the pattern of caveolin expression in benign prostatic epithelium and androgen-sensitive human prostate cancer is consistent with tumor suppressive activity.


Assuntos
Caveolinas , Di-Hidrotestosterona/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Membrana/genética , Neoplasias da Próstata/genética , Animais , Western Blotting , Castração/veterinária , Caveolina 1 , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Nus , Próstata/citologia , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Distribuição Aleatória , Células Tumorais Cultivadas
6.
Endocrinology ; 136(1): 262-8, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7828539

RESUMO

Nerve growth factor-beta (NGF beta) and a NGF beta-immunoreactive protein derived from human prostatic stromal cell secretory protein (hPS) have been shown to stimulate the growth of prostate epithelial cells. An NGF beta-immunoreactive protein has been localized to the stroma of human prostate tissues, and a low affinity NGF receptor (gp75NGFR) has been localized to the adjacent epithelia, consistent with the paracrine regulation of prostate growth. Interestingly, gp75NGFR is progressively lost during neoplastic progression of the human prostate. In this report we have characterized the expression of the signal-transducing component of the NGF receptor, the Trk tyrosine receptor kinase, in prostate epithelial cells that bind exogenous NGF beta and an endogenous NGF beta-immunoreactive protein in hPS. In this context, a pan-Trk antibody that recognizes all of the members of the Trk receptor family (TrkA, TrkB, and TrkC) specifically localized expression of the Trk receptor to the epithelial component of normal prostate tissue, benign prostatic hyperplasia, and adenocarcinoma tissue. The binding of [125I]NGF beta to the surface of primary cultures of human prostate epithelia and the TSU-pr1 human metastatic prostate tumor cell line was displaced with either excess cold NGF beta or hPS, whereas binding was not displaced by epidermal growth factor or platelet-derived growth factor. Scatchard plot analysis of [125I]NGF beta binding to these cells identified a low affinity binding site (Kd = 1.9 x 10(-9) M) and a high affinity binding site (Kd = 1.8 x 10(-11) M) on the primary prostate epithelia, whereas only a high affinity binding site (Kd = 1.3 x 10(-11) M) was observed on the TSU-pr1 tumor cells. Stimulation of TSU-pr1 cells with either NGF beta or hPS induced tyrosine phosphorylation of Trk proteins, whereas no phosphorylation was evident in untreated cells, cells treated with hPS immunoprecipitated with anti-NGF beta antibody, or brain-derived neurotrophic factor- and neurotrophin-3-treated cells. The Trk protein was also observed in these cells by immunoblot analysis with pan-Trk antibody. These results demonstrate a functional Trk receptor in the epithelia of the human prostate that is responsive to exogenous NGF beta and an endogenous NGF beta-immunoreactive protein in hPS, thereby supporting the concept of the paracrine regulation of growth in the human prostate via a stromal neurotrophin-epithelial Trk receptor interaction.


Assuntos
Próstata/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Ligação Competitiva , Epitélio/metabolismo , Epitélio/patologia , Humanos , Imuno-Histoquímica , Ligantes , Masculino , Fosforilação , Próstata/patologia , Receptor trkA , Distribuição Tecidual , Células Tumorais Cultivadas
7.
Cancer Res ; 53(6): 1416-20, 1993 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-8443820

RESUMO

The migration of three human prostate tumor epithelial cell lines (TSU-pr1, PC-3, DU-145) in response to secreted protein from a human prostate stromal cell line was investigated by using the modified blind-well Boyden chamber assay. Migrated cells were quantified by spectrophotometrically measuring the concentration of crystal violet stain extracted from their nuclei. Cell number was correlated linearly with the concentration of extracted crystal violet stain. All three tumor cell lines showed intrinsic migratory ability in the absence of chemoattractants, such that approximately 1-7% of plated cells migrated across the filter of the Boyden chambers during a 5-h incubation period. Prostate tumor cell migration was significantly enhanced (3-13-fold) in response to stromal cell secretory protein in a dose-dependent manner, whereas bovine serum albumin had no effect on stimulating tumor cell migration. Immunoprecipitation of the stromal cell secreted protein with a nerve growth factor antibody partially and significantly reduced its stimulatory activity for tumor cell migration. A Zigmond-Hirsch matrix assay of tumor cell migration in response to various concentration gradients of stromal cell secreted protein demonstrated both chemotaxis and chemokinesis by all three cell lines. These results are consistent with the stromal cell secretory protein stimulation of chemokinetic tumor cell migration through the capsule of the prostate. Outside of the prostate gland metastasis of tumor cells may occur by chemotaxis to preferential sites containing chemoattractants similar to or related to maintenance factors that can substitute for components of stromal cell secretory protein.


Assuntos
Proteínas de Transporte/farmacologia , Quimiotaxia , Fatores de Crescimento Neural/farmacologia , Próstata/citologia , Neoplasias da Próstata/patologia , Proteínas Secretadas pela Próstata , Movimento Celular/efeitos dos fármacos , Humanos , Masculino , Espectrofotometria , Células Estromais/fisiologia , Células Tumorais Cultivadas
8.
Cancer Res ; 52(19): 5403-6, 1992 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-1382843

RESUMO

In the human prostate, a low affinity (p75) nerve growth factor (NGF) receptor (NGF-R) localizes to the epithelia while a NGF-like protein localizes to the stroma. This NGF-like ligand, derived from prostate stromal cell cultures, has been shown to participate in paracrine mediated growth of a human tumor epithelial cell line (TSU-prl) in vitro. In order to investigate the role of the NGF-R in neoplastic growth we have examined the expression of the NGF-R in normal prostate tissues, benign prostatic hyperplasia tissues, adenocarcinoma tissues, and four metastatic tumor cell lines of the human prostate. In primary epithelial cell cultures of normal human prostate the p75 NGF-R was localized by immunocytochemistry to cytoplasmic vesicles. Furthermore, Western blot analysis of the NGF-R in subcellular fractions of normal prostate tissue identified an M(r) 75,000 immunoreactive protein in the microsomal fraction under nonreducing conditions of sodium dodecyl sulfatepolyacrylamide gel electrophoresis. However, microsomal preparations of five prostatic adenocarcinoma and five benign prostatic hyperplasia specimens showed varying immunoreactivity among samples, all of which expressed less of the p75 NGF-R than the normal tissue. Interestingly, microsomal preparations of the human prostatic epithelial cell lines, TSU-prl, DU-145, PC-3, and LNCaP did not show NGF-R expression by immunoblot analysis. Hence, expression of the p75 NGF-R in normal prostate tissue, partial loss of NGF-R expression in benign and malignant prostate tissue, and complete loss of NGF-R expression in the four metastatic tumor cell lines, suggests an inverse association of p75 NGF-R expression with the neoplastic progression of the human prostate.


Assuntos
Adenocarcinoma/ultraestrutura , Próstata/ultraestrutura , Hiperplasia Prostática/patologia , Neoplasias da Próstata/ultraestrutura , Receptores de Fator de Crescimento Neural/análise , Imunofluorescência , Humanos , Immunoblotting , Imuno-Histoquímica , Masculino , Neoplasias da Próstata/secundário , Receptores de Fator de Crescimento Neural/fisiologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA