Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Cancer ; 19(1): 1114, 2019 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-31729966

RESUMO

We investigated the association of LHR expression in epithelial ovarian cancer (OC) with clinical and pathologic characteristics of patients. LHR expression was examined immunohistochemically using tissue microarrays (TMAs) of specimens from 232 OC patients. Each sample was scored quantitatively evaluating LHR staining intensity (LHR-I) and percentage of LHR (LHR-P) staining cells in tumor cells examined. LHR-I was assessed as no staining (negative), weak (+ 1), moderate (+ 2), and strong positive (+ 3). LHR-P was measured as 1 to 5, 6 to 50% and >  50% of the tumor cells examined. Positive LHR staining was found in 202 (87%) patients' tumor specimens and 66% patients had strong intensity LHR expression. In 197 (85%) of patients, LHR-P was measured in > 50% of tumor cells. LHR-I was significantly associated with pathologic stage (p = 0.007). We found that 72% of stage III or IV patients expressed strong LHR-I in tumor cells. There were 87% of Silberberg's grade 2 or 3 patients compared to 70% of grade 1 patients with LHR expression observed in > 50% of tumor cells, p = 0.037. Tumor stage was significantly associated with overall survival and recurrence free survival, p < 0.001 for both analyses, even after adjustment for age, tumor grade and whether patient had persistent disease after therapy or not. Our study demonstrates that LHR is highly expressed in the majority of OC patients. Both LHR-I and LHR-P are significantly associated with either the pathologic stage or tumor grade.


Assuntos
Carcinoma Epitelial do Ovário/metabolismo , Neoplasias Ovarianas/metabolismo , Receptores do LH/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/biossíntese , Carcinoma Epitelial do Ovário/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Neoplasias Ovarianas/patologia
2.
Clin Genitourin Cancer ; 15(6): 742-749, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28668277

RESUMO

BACKGROUND: AEZS-108 (zoptarelin doxorubicin) is a cytotoxic hybrid molecule consisting of doxorubicin covalently coupled with a luteinizing hormone-releasing hormone (LHRH) analogue, which selectively targets doxorubicin to tumor cells expressing LHRH receptors. We report the clinical efficacy of AEZS-108 in a phase II trial in men with metastatic castrate-resistant prostate cancer who had disease progression after taxane-based chemotherapy. PATIENTS AND METHODS: Patients received AEZS-108 210 mg/m2 intravenously every 3 weeks. The primary end point was clinical benefit defined as nonprogression at 12 weeks with no dose-limiting toxicities (DLTs) or other toxicities requiring termination of treatment. Secondary end points included response rate, pain response, progression-free survival (PFS), and overall survival (OS). Circulating tumor cells (CTCs) were captured and tested for LHRH receptors, as well as for internalization of AEZS-108 using autofluorescence. RESULTS: Twenty-five patients were enrolled; 20 patients had at least 1 measurable lesion at baseline. Patients received a median of 5 cycles (range, 1-9) and 44% of patients received at least 6 cycles with 2 patients who completed ≥ 8 cycles. Considering clinical benefits, 13 patients (52%) remained progression-free at 12 weeks with no DLT or other toxicities requiring termination of treatment. For clinical response according to Response Evaluation Criteria in Solid Tumors version 1.1 criteria, 1 patient (4%) experienced a confirmed partial response (PR) within 12 weeks, 14 patients (56%) had stable disease (SD), and 8 patients (32%) had disease progression. For maximal prostate-specific antigen (PSA) response, 1 patient (4%) experienced a confirmed PR within 12 weeks, 21 patients (84%) had SD, and 3 patients (12%) had disease progression as denoted by their best PSA response. Pain improved in 13 (59%) patients. The median PFS was 3.8 months (95% confidence interval [CI], 2.1-4.4), and median OS was 6.0 months (95% CI, 4.2-10.1) with a median follow-up of 16.1 months (range, 3.2-36.1). Baseline CTC enumeration was an independent predictor of OS but not PFS. CONCLUSION: AEZS-108 showed activity in patients who were pretreated, a subset typically very difficult to treat, and maintained an acceptable safety profile.


Assuntos
Doxorrubicina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/análogos & derivados , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Esquema de Medicação , Hormônio Liberador de Gonadotropina/administração & dosagem , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Células Neoplásicas Circulantes/efeitos dos fármacos , Células Neoplásicas Circulantes/metabolismo , Antígeno Prostático Específico/metabolismo , Neoplasias de Próstata Resistentes à Castração/metabolismo , Receptores do LH/metabolismo , Análise de Sobrevida , Taxoides/uso terapêutico , Resultado do Tratamento
3.
Prostate ; 75(2): 141-50, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25393079

RESUMO

BACKGROUND: The importance of androgen signaling in prostate cancer (PC) is well described and prostate cancer cells retain the ability to directly synthesize androgens. Luteinizing hormone (LH) can induce expression of steroidogenic enzymes and trigger androgen production, but the regulation of this process is not well-described. Here, we explored the impact of silencing LH receptor (LHR) silencing on androgen synthesis and on several relevant signaling pathways in PC. METHODS: LHR mRNA and protein expression was evaluated in LNCaP PC cells treated with LHR-siRNA. MTS assay was used to measure the effect of LHR-siRNA on proliferation in LNCaP and 22RV1 PC cells. Treated LNCaP and LAPC-3 cells were also assayed for differences in androgen synthesis and expression of steroidogenic enzymes, PSA, AR, and critical signaling molecules including PKA, ERK1/2, PI3K, AKT2, and HER2. RESULTS: We confirmed that functional LHR is expressed in both androgen-sensitive and castrate-resistant PC specimens. Treatment with LHR-siRNA effectively silenced LHR gene and protein expression and prevented LH-mediated proliferation and androgen synthesis in prostate cancer cells. LHR silencing also downregulated expression of AR, PSA, PKA, ERK1/2, PI3K, AKT2, and HER2. CONCLUSION: Collectively, these data demonstrate that silencing LHR expression suppresses androgen synthesis and signaling and the LH-LHR pathway may represent a viable therapeutic strategy in PC.


Assuntos
Inativação Gênica/fisiologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptores do LH/biossíntese , Receptores do LH/genética , Transdução de Sinais/fisiologia , Androgênios/biossíntese , Animais , Inativação Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias da Próstata/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
4.
Clin Cancer Res ; 20(24): 6277-83, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25278449

RESUMO

PURPOSE: AEZS-108, formerly AN-152, is a cytotoxic hybrid molecule consisting of a luteinizing hormone-releasing hormone (LHRH) agonist moiety covalently coupled to doxorubicin, allowing it to deliver doxorubicin selectively to cells expressing LHRH receptors. LHRH receptors are expressed on the cell membrane of many tumors, including prostate cancer. This phase I study determined the maximum tolerated dose (MTD) of AEZS-108 in men with taxane- and castration-resistant prostate cancer (CRPC) while providing additional information on the safety profile and efficacy of this agent. EXPERIMENTAL DESIGN: AEZS-108 was administered as an intravenous infusion every 21 days until progression or unacceptable toxicity in cohorts of 3 or 6 patients until the MTD was reached. Blood was collected for capture of circulating tumor cells (CTC) to visualize internalization of AEZS-108, an autofluorescent molecule. RESULTS: The MTD of AEZS-108 in this cohort was 210 mg/m(2), which was lower than that seen in a phase I study conducted in women with endometrial or ovarian cancers. The dose-limiting toxicity was persistent neutropenia. Three patients had a PSA response with an additional 10 patients maintaining PSA stable disease. Of the 10 patients evaluable by RECIST criteria, 9 achieved stable disease. AEZS-108 internalization in CTCs was routinely visualized using its autofluorescence. CONCLUSION: These findings show that AEZS-108 has an acceptable safety profile and a signal of efficacy, lowering PSA in heavily pretreated patients with prostate cancer, and that internalization of AEZS-108 in prostate cancer CTCs may be a viable pharmacodynamic marker. A phase II study in men with prostate cancer is ongoing.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Doxorrubicina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Hormônio Liberador de Gonadotropina/análogos & derivados , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos Hormonais/administração & dosagem , Antineoplásicos Hormonais/efeitos adversos , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Hidrocarbonetos Aromáticos com Pontes/uso terapêutico , Doxorrubicina/administração & dosagem , Doxorrubicina/efeitos adversos , Doxorrubicina/uso terapêutico , Hormônio Liberador de Gonadotropina/administração & dosagem , Hormônio Liberador de Gonadotropina/efeitos adversos , Hormônio Liberador de Gonadotropina/uso terapêutico , Humanos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Neoplasia Residual , Neoplasias de Próstata Resistentes à Castração/mortalidade , Neoplasias de Próstata Resistentes à Castração/patologia , Retratamento , Taxoides/farmacologia , Taxoides/uso terapêutico , Resultado do Tratamento
5.
Clin Genitourin Cancer ; 11(4): 416-22, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24099865

RESUMO

BACKGROUND: Although new androgen-targeted therapies offer prolonged survival in metastatic castration-resistant prostate cancer (CRPC), most men still face progressive disease and require additional therapy. Oxaliplatin and pemetrexed have each shown modest activity in the treatment of CRPC. Given their favorable nonoverlapping toxicity profiles, we studied them in combination. PATIENTS AND METHODS: Men with CRPC whose disease had progressed on 1 or 2 previous chemotherapy regimens, including a taxane, were eligible. All participants received oxaliplatin 120 mg/m(2) and pemetrexed 500 mg/m(2) intravenously every 21 days. The primary end point was response rate; objective responses were determined using Response Evaluation Criteria in Solid Tumors (RECIST), version 1.0, criteria and the Prostate Cancer Working Group (1999) criteria. Secondary end points included progression-free survival and OS. RESULTS: Forty-seven men received a median of 6 cycles (range, 1-21). The overall response rate was 30% (95% confidence interval [CI], 18%-45%), including 10 men with RECIST responses of the 40 who had measurable disease (25%). Overall, 64% had a prostate-specific antigen (PSA) decline and 74% of men had clinical disease control (partial response or stable disease as their best response). Median progression-free survival was 5.8 months (95% CI, 3.8-7.6), with a median OS of 11.9 months. Six of 15 evaluable patients (40%) experienced a pain response. Nineteen patients (40%) experienced a grade 3 or 4 hematologic toxicity, and 16 (34%) experienced a grade 3 nonhematologic toxicity. One patient died while participating in the study. CONCLUSION: Combination oxaliplatin and pemetrexed (PemOx) is an effective and tolerable second- or third-line treatment for men with CRPC.


Assuntos
Antineoplásicos/uso terapêutico , Glutamatos/uso terapêutico , Guanina/análogos & derivados , Compostos Organoplatínicos/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Idoso , Antineoplásicos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Hidrocarbonetos Aromáticos com Pontes/uso terapêutico , Intervalo Livre de Doença , Glutamatos/efeitos adversos , Guanina/efeitos adversos , Guanina/uso terapêutico , Humanos , Masculino , Compostos Organoplatínicos/efeitos adversos , Oxaliplatina , Dor/tratamento farmacológico , Manejo da Dor , Pemetrexede , Antígeno Prostático Específico/sangue , Neoplasias de Próstata Resistentes à Castração/mortalidade , Sobrevida , Taxoides/uso terapêutico , Resultado do Tratamento
6.
Anticancer Res ; 31(11): 3897-901, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22110216

RESUMO

BACKGROUND/AIM: The role of neuroendocrine (NE) cells in prostate cancer biology remains unclear. We previously reported a large difference in NE expression in benign prostate tissue among men of different ethnicities; African-American men had significantly fewer NE cells compared to all other groups. This report describes NE expression in malignant prostate tissue. PATIENTS AND METHODS: Paraffin-embedded tissue from 180 men who underwent radical prostatectomy at the University of Southern California between 1983 and 2003 was stained using standard immunohistochemistry technique for chromogranin A (ChrA), serotonin (Ser) and synaptophysin (Syn). There were 39 specimens from African-American patients, 39 Asian, 57 Hispanic and 45 non-Hispanic White. Staining intensity and the percentage of cells positive were determined by the automated cellular imaging system. Results were analyzed by univariate and multivariate general linear regression models. RESULTS: There were significant differences in staining intensity for all markers between ethnic groups in univariate analysis. NE expression, judged by ChrA intensity, was highest in Hispanic patients, compared to non-Hispanic Whites and African-Americans. A similar pattern was observed for Syn and Ser. In multivariate analysis, controlling for age, Gleason score, PSA and stage, the differences in ChrA, Syn and Ser remained highly significant. Hispanic men had higher ChrA expression levels than African-Americans and non-Hispanic Whites (p=0.0077 and 0.0038, respectively); the p-values for the comparison were both <0.0001 for Ser. Both Hispanic and Asian patients had higher intensity Ser expression than African-American and Non-Hispanic Whites patients, with all p-values <0.018. CONCLUSION: As already shown in benign prostate tissue, we identified significant differences in NE expression among prostate cancer tissues from men of different ethnic backgrounds. The clinical impact of these differences in NE expression warrants exploration.


Assuntos
Cromogranina A/metabolismo , Células Neuroendócrinas/metabolismo , Neoplasias da Próstata/etnologia , Neoplasias da Próstata/metabolismo , Serotonina/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Negro ou Afro-Americano/estatística & dados numéricos , Idoso , Povo Asiático/estatística & dados numéricos , California , Hispânico ou Latino/estatística & dados numéricos , Humanos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Células Neuroendócrinas/patologia , Prostatectomia , Neoplasias da Próstata/patologia , Sinaptofisina , População Branca/estatística & dados numéricos
7.
Prostate ; 71(8): 892-8, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21456071

RESUMO

BACKGROUND: Recent data has shown that prostate cancer (PCA) cells are capable of producing testosterone directly from cholesterol, which may contribute to the development of castration resistance. While up-regulation of steroidogenic enzymes has been previously described during castration-resistant prostate cancer (CRPC) progression, regulation of this process is poorly defined. These data examine the role of luteinizing hormone (LH) in the regulation of steroidogenic machinery in PCA cells. METHODS: PCA cell lines LNCaP, C4-2B, and 22RV1 were exposed to LH. Gene expression was quantified using real-time PCR and protein expression was characterized with standard Western blot analysis. Steroid analysis was performed using radioimmunoassay (RIA). Cell viability was measured using an MTS viability assay. RESULTS: Androgen-sensitive (LNCaP) and -independent PCA cells (C4-2B and 22RV1) express both mRNA and protein for LH and LH receptor (LHR). Exposure of these cells to LH for 4 hr increased the expression of several steroidogenic genes. Exposure for 10 days resulted in the increase of additional genes. At both time points, the upregulation of these genes was dose-dependent. This was mirrored by an increase in the expression of several key steroidogenic enzymes, including StAR, CYB5B, CYP11A, and 3ßHSD. LH stimulated the production of progesterone and testosterone in LNCaP cells as measured by RIA. We have also demonstrated that treatment of LNCaP cells with LH enhanced their viability. CONCLUSIONS: Our data show that LH-mediated activation of LHR significantly up-regulates the expression of genes and enzymes required for steroidogenesis and increases steroid production in PCA cells.


Assuntos
Carcinoma/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hormônio Luteinizante/farmacologia , Progesterona/biossíntese , Neoplasias da Próstata/metabolismo , Testosterona/biossíntese , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Humanos , Masculino , Receptores do LH/biossíntese , Regulação para Cima
8.
Cancer Res ; 71(8): 2848-57, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21467168

RESUMO

Glucose-regulated protein 78 (GRP78)/BiP is a multifunctional protein which plays a major role in endoplasmic reticulum (ER) protein processing, protein quality control, maintaining ER homeostasis, and controlling cell signaling and viability. Previously, using a transgene-induced mammary tumor model, we showed that Grp78 heterozygosity impeded cancer growth through suppression of tumor cell proliferation and promotion of apoptosis and the Grp78(+/-) mice exhibited dramatic reduction (70%) in the microvessel density (MVD) of the endogenous mammary tumors, while having no effect on the MVD of normal organs. This observation suggests that GRP78 may critically regulate the function of the host vasculature within the tumor microenvironment. In this article, we interrogated the role of GRP78 in the tumor microenvironment. In mouse tumor models in which wild-type (WT), syngeneic mammary tumor cells were injected into the host, we showed that Grp78(+/-) mice suppressed tumor growth and angiogenesis during the early phase but not during the late phase of tumor growth. Growth of metastatic lesions of WT, syngeneic melanoma cells in the Grp78(+/-) mice was potently suppressed. We created conditional heterozygous knockout of GRP78 in the host endothelial cells and showed severe reduction of tumor angiogenesis and metastatic growth, with minimal effect on normal tissue MVD. Furthermore, knockdown of GRP78 expression in immortalized human endothelial cells showed that GRP78 is a critical mediator of angiogenesis by regulating cell proliferation, survival, and migration. Our findings suggest that concomitant use of current chemotherapeutic agents and novel therapies against GRP78 may offer a powerful dual approach to arrest cancer initiation, progression, and metastasis.


Assuntos
Proteínas de Choque Térmico/genética , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/genética , Animais , Apoptose/fisiologia , Processos de Crescimento Celular/fisiologia , Movimento Celular/fisiologia , Chaperona BiP do Retículo Endoplasmático , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Feminino , Heterozigoto , Humanos , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Metástase Neoplásica , Neovascularização Patológica/genética , Microambiente Tumoral
9.
Clin Cancer Res ; 16(18): 4675-80, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20670943

RESUMO

PURPOSE: In addition to their expression on pituitary cells, receptors for luteinizing hormone-releasing hormone (LH-RH) are found on most prostate cancer cells. These tumoral LH-RH receptors mediate the direct cytotoxic effects of LH-RH analogs and are potential therapeutic targets. Although pituitary LH-RH receptors are downregulated following prolonged exposure to LH-RH agonists, there is no evidence that tumoral receptors behave in a similar manner. To better characterize expression of tumoral LH-RH receptors, specimens of prostate cancer from various cohorts of patients were analyzed. EXPERIMENTAL DESIGN: Surgical specimens were obtained from untreated patients with prostate cancer and from patients with metastatic castration-resistant prostate cancer previously treated with bilateral orchiectomy. To address the possibility of receptor downregulation, two additional cohorts of patients who had been previously treated with LH-RH agonists were included. One group received neoadjuvant therapy prior to prostatectomy, and the other group was treated for metastatic disease with LH-RH agonists and, at progression, required palliative resection of the prostate. Lymph node metastases from previously untreated patients were subjected to similar analysis. RESULTS: Expression of LH-RH receptors was found in most specimens. The relative expression of LH-RH receptor mRNA in untreated patients was greater in patients whose tumor had received a Gleason score <8. CONCLUSIONS: LH-RH receptor expression persisted despite prolonged exposure to LH-RH agonists. These findings support the concept of targeting cytotoxic LH-RH analogs to prostatic LH-RH receptors, using these receptors to gain entry into cancer cells to deliver a hybridized cytotoxic moiety for the treatment of prostate cancer.


Assuntos
Adenocarcinoma/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Receptores LHRH/agonistas , Receptores LHRH/genética , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma/cirurgia , Quimioterapia Adjuvante , Regulação Neoplásica da Expressão Gênica , Hormônio Liberador de Gonadotropina/análogos & derivados , Humanos , Metástase Linfática , Masculino , Orquiectomia , Prostatectomia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia , Receptores LHRH/metabolismo , Falha de Tratamento
10.
Prostate ; 70(15): 1692-700, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20564320

RESUMO

BACKGROUND: Androgens stimulate the expression of vascular endothelial growth factor (VEGF) through activation of hypoxia inducible factor (HIF). These genes play a major role in cancer angiogenesis. This study assesses the relationship among expression levels for the androgen receptor (AR), HIF1a, VEGF-A, and VEGF-C genes in human prostate cancer tissue and their impact on prostate cancer outcomes. It also examines the impact of pre-operative androgen deprivation therapy (ADT) on the expression of these genes. METHODS: Radical prostatectomy specimens were obtained from 138 patients with D1 prostate cancer from the University of Southern California prostatectomy database; 30% received pre-operative and 23% received post-operative ADT. Gene expression levels were determined by quantitative real-time PCR. Specimens were stratified into three groups for each gene based on expression levels, and groups were compared for clinical outcomes (PSA and clinical recurrence, overall survival). RESULTS: There was a significant correlation in expression levels amongst all genes. Patients treated with pre-operative ADT had significantly lower HIF1a expression, mean 2.64 (CI 2.34-2.94) than patients not treated, mean 3.25 (CI 2.97-3.53, P = 0.006), adjusting for age, PSA, Gleason score, and stage. Higher VEGF-A expression was significantly associated with better overall survival (HR 0.49, P = 0.015). The risk of developing clinical recurrence was significantly lower with higher VEGF-C expression (HR 0.4, P = 0.014). CONCLUSIONS: Significant correlation was noted among AR, HIF1a, VEGF-A, and VEGF-C. This study shows that ADT is associated with lower HIF1a gene expression in human prostate cancer tissue and documents prognostic value for VEGF-A and VEGF-C expression levels.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/genética , Idoso , Antagonistas de Androgênios/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Hormônio-Dependentes/genética , Neoplasias Hormônio-Dependentes/patologia , Neoplasias Hormônio-Dependentes/terapia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Prognóstico , Modelos de Riscos Proporcionais , Prostatectomia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Androgênicos/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatísticas não Paramétricas , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator C de Crescimento do Endotélio Vascular/biossíntese
11.
Prostate ; 69(1): 12-23, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18814146

RESUMO

BACKGROUND: Neuroendocrine (NE) cells are present in both normal prostate and prostate cancer. In addition, NE differentiation can be induced by various factors, such as IL-6, in vitro and in vivo. However, the mechanism of this differentiation and the role of NE cells in prostate cancer are not well understood. In this study, we evaluated the gene expression and analyzed the pathways in prostate cancer cells exposed to various NE differentiation inducing factors in vitro. METHODS: Gene expression signatures between control LNCaP cells and each treatment induced NE cell line were compared using Affymetrix GeneChip with network and pathway analysis. RESULTS: All treatments were able to transdifferentiate LNCaP cells into NE phenotype as shown by morphology changes and NE marker measurements. Of the 54,675 oligonucleotide-based probe sets in microarray, 44,975 were mapped into the Ingenuity Pathway Analysis database and were filtered according to the t-test P value. At P < 0.002, the number of genes that were differentially expressed included 302 of the IL-6 treated cells, 201 of genistein, 233 of epinephrine, and 191 of the charcoal stripped serum ones. A pooled data approach also showed 346 differentially expressed genes at the same P value. Gene ontology analysis showed that cancer-related function had the highest significance. CONCLUSIONS: Despite some overlap, each NE transdifferentiation inducing treatment was associated with a changed expression of a unique set of genes, and such gene profiling may help to elucidate the molecular mechanisms involved in NE transdifferentiation of prostate cancer cells.


Assuntos
Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Sistemas Neurossecretores/citologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Anticarcinógenos/farmacologia , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Bases de Dados Genéticas , Genisteína/farmacologia , Humanos , Interleucina-6/farmacologia , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fenótipo , Neoplasias da Próstata/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Alcohol Clin Exp Res ; 32(11): 1947-53, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18782341

RESUMO

BACKGROUND: Alcohol consumption is associated with increased iron stores. In sub-Saharan Africa, high dietary ionic iron and the ferroportin Q248H allele have also been implicated in iron accumulation. We examined the associations of ferroportin Q248H, alcohol and dietary iron with serum ferritin, aspartate aminotransaminase (AST) and alanine aminotransaminase (ALT) concentrations in African-Americans. METHODS: Inner-city African-Americans (103 men, 40 women) were recruited from the community according to reported ingestion of >4 alcoholic drinks/d or <2/wk. Typical daily heme iron, nonheme iron and alcohol were estimated using University of Hawaii's multiethnic dietary questionnaire. Based on dietary questionnaire estimates we established categories of < versus > or =56 g alcohol/d, equivalent to 4 alcoholic drinks/d assuming 14 g alcohol per drink. RESULTS: Among 143 participants, 77% drank <56 g alcohol/d and 23%> or =56 g/d as estimated by the questionnaire. The prevalence of ferroportin Q248H was 23.3% with alcohol >56 g/d versus 7.5% with lower amounts (p = 0.014). Among subjects with no history of HIV disease, serum ferritin concentration had positive relationships with male gender (p = 0.041), alcohol consumption (p = 0.021) and ALT concentration (p = 0.0001) but not with dietary iron intake or ferroportin Q248H. Serum AST and ALT concentrations had significant positive associations with male gender and hepatitis C seropositivity but not with alcohol or dietary iron intake or ferroportin Q248H. CONCLUSIONS: Our findings suggest a higher prevalence of ferroportin Q248H with greater alcohol consumption, and this higher prevalence raises the possibility that the allele might ameliorate the toxicity of alcohol. Our results suggest that alcohol but not dietary iron contributes to higher body iron stores in African-Americans. Studies with larger numbers of participants are needed to further clarify the relationship of ferroportin Q248H with the toxicity of alcohol consumption.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Consumo de Bebidas Alcoólicas/metabolismo , Alelos , Negro ou Afro-Americano/genética , Proteínas de Transporte de Cátions/genética , Ferritinas/sangue , Ferro da Dieta/metabolismo , Adulto , Alanina Transaminase/sangue , Consumo de Bebidas Alcoólicas/etnologia , Aspartato Aminotransferases/sangue , Feminino , Ferritinas/metabolismo , Inquéritos Epidemiológicos , Hemeproteínas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Estatísticos , Mutação/genética , Análise de Regressão , População Urbana
13.
Am J Physiol Gastrointest Liver Physiol ; 295(3): G512-21, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18599584

RESUMO

One prime feature of alcoholic liver disease (ALD) is iron accumulation in hepatic macrophages/Kupffer cells (KC) associated with enhanced NF-kappaB activation. Our recent work demonstrates a peroxynitrite-mediated transient rise in intracellular labile iron (ILI) as novel signaling for endotoxin-induced IKK and NF-kappaB activation in rodent KC. The present study investigated the mechanism of KC iron accumulation and its effects on ILI response in experimental ALD. We also tested ILI response in human blood monocytes. Chronic alcohol feeding in rats results in increased expression of transferrin (Tf) receptor-1 and hemochromatosis gene (HFE), enhanced iron uptake, an increase in nonheme iron content, and accentuated ILI response for NF-kappaB activation in KC. Ex vivo treatment of these KC with an iron chelator abrogates the increment of iron content, ILI response, and NF-kappaB activation. The ILI response is evident in macrophages derived from human blood monocytes by PMA treatment but not in vehicle-treated monocytes, and this differentiation-associated phenomenon is essential for maximal TNF-alpha release. PMA-induced macrophages load iron dextran and enhance ILI response and TNF-alpha release. These effects are reproduced in KC selectively loaded in vivo with iron dextran in mice and more importantly aggravate experimental ALD. Our results suggest enhanced iron uptake as a mechanism of KC iron loading in ALD and demonstrate the ILI response as a function acquired by differentiated macrophages in humans and as a priming mechanism for ALD.


Assuntos
Fígado Gorduroso Alcoólico/metabolismo , Ferro/metabolismo , Células de Kupffer/metabolismo , Fígado/metabolismo , Animais , Proteínas de Transporte de Cátions/metabolismo , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Etanol , Fígado Gorduroso Alcoólico/etiologia , Fígado Gorduroso Alcoólico/patologia , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Quelantes de Ferro/farmacologia , Complexo Ferro-Dextran , Células de Kupffer/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Ratos , Ratos Wistar , Receptores da Transferrina/metabolismo , Transdução de Sinais , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
14.
Cancer Res ; 68(2): 498-505, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18199545

RESUMO

The unfolded protein response (UPR) is an evolutionarily conserved mechanism that activates both proapoptotic and survival pathways to allow eukaryotic cells to adapt to endoplasmic reticulum (ER) stress. Although the UPR has been implicated in tumorigenesis, its precise role in endogenous cancer remains unclear. A major UPR protective response is the induction of the ER chaperone GRP78/BiP, which is expressed at high levels in a variety of tumors and confers drug resistance in both proliferating and dormant cancer cells. To determine the physiologic role of GRP78 in in situ-generated tumor and the consequence of its suppression on normal organs, we used a genetic model of breast cancer in the Grp78 heterozygous mice where GRP78 expression level was reduced by about half, mimicking anti-GRP78 agents that achieve partial suppression of GRP78 expression. Here, we report that Grp78 heterozygosity has no effect on organ development or antibody production but prolongs the latency period and significantly impedes tumor growth. Our results reveal three major mechanisms mediated by GRP78 for cancer progression: enhancement of tumor cell proliferation, protection against apoptosis, and promotion of tumor angiogenesis. Importantly, although partial reduction of GRP78 in the Grp78 heterozygous mice substantially reduces the tumor microvessel density, it has no effect on vasculature of normal organs. Our findings establish that a key UPR target GRP78 is preferably required for pathophysiologic conditions, such as tumor proliferation, survival, and angiogenesis, underscoring its potential value as a novel therapeutic target for dual antitumor and antiangiogenesis activity.


Assuntos
Proliferação de Células , Proteínas de Choque Térmico/fisiologia , Neoplasias Mamárias Experimentais/patologia , Chaperonas Moleculares/fisiologia , Neovascularização Patológica/genética , Animais , Formação de Anticorpos/genética , Apoptose/genética , Caspases/genética , Sobrevivência Celular , Chaperona BiP do Retículo Endoplasmático , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico/genética , Heterozigoto , Masculino , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares/genética , Fator de Transcrição CHOP/genética , Transgenes/fisiologia , Carga Tumoral/genética
15.
Am J Physiol Gastrointest Liver Physiol ; 294(1): G39-49, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18006602

RESUMO

Activation of hepatic stellate cells (HSC), a key event in liver fibrosis, is caused by diminished adipogenic transcription. This study investigated whether Wnt signaling contributes to "antiadipogenic" activation of HSC and liver fibrogenesis. Culture-activated HSC from normal rats and HSC from cholestatic rat livers were examined for expression of Wnt, Frizzled (Fz) receptors, and coreceptors by quantitative PCR. Wnt signaling was assessed by nuclear beta-catenin and T cell factor (TCF) promoter activity. Dickkopf-1 (Dkk-1), a Wnt coreceptor antagonist, was transduced by an adenoviral vector to assess the effects of Wnt antagonism on culture activation of HSC and cholestatic liver fibrosis in mice. Messenger RNA for canonical (Wnt3a and 10b) and noncanonical (Wnt4 and 5a) Wnt genes, Fz-1 and 2, and coreceptors [low-density lipoprotein-receptor-related protein (LRP)6 and Ryk] are increased approximately 3-12-fold in culture-activated HSC compared with quiescent HSC. The nuclear beta-catenin level and TCF DNA binding are markedly increased in activated HSC. TCF promoter activity is stimulated with Wnt1 but inhibited by Chibby, a protein that blocks beta-catenin interaction with TCF, and by Dkk-1. Dkk-1 enhances peroxisome proliferator-activated receptor-gamma (PPARgamma)-driven PPAR response element (PPRE) promoter activity, a key adipogenic transcriptional parameter, abrogates agonist-stimulated contraction, and restores HSC quiescence in culture. High expression of Dkk-1 increases apoptosis of cultured HSC. Expression of Wnt and Fz genes is also induced in HSC isolated from experimental cholestatic liver fibrosis, and Dkk-1 expression ameliorates this form of liver fibrosis in mice. These results demonstrate antiadipogenic Wnt signaling in HSC activation and therapeutic potential of Wnt antagonism for liver fibrosis.


Assuntos
Receptores Frizzled/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Cirrose Hepática Biliar/metabolismo , Fígado/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Animais , Apoptose , Linhagem Celular , Núcleo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Ducto Colédoco/cirurgia , Modelos Animais de Doenças , Receptores Frizzled/genética , Terapia Genética/métodos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Ligadura , Fígado/patologia , Cirrose Hepática Biliar/genética , Cirrose Hepática Biliar/patologia , Cirrose Hepática Biliar/prevenção & controle , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de LDL/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição TCF/metabolismo , Fatores de Tempo , Transcrição Gênica , Transfecção , Regulação para Cima , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/genética , beta Catenina/metabolismo
16.
J Biol Chem ; 282(8): 5582-8, 2007 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-17172471

RESUMO

We recently discovered a novel signaling phenomenon involving a rapid and transient rise in intracellular low molecular weight iron complex(es) in activation of IkappaB kinase (IKK) in hepatic macrophages. We also showed direct treatment with ferrous iron substitutes for this event to activate IKK. The present study used this model to identify upstream kinases responsible for IKK activation. IKK activation induced by iron is abrogated by overexpression of a dominant negative mutant (DN) for transforming growth factor beta-activated kinase-1 (TAK1), NF-kappaB-inducing kinase, or phosphatidylinositol 3-kinase (PI3K) and by treatment with the mitogen-activated protein kinase (MAPK) kinase-1 (MEK1) inhibitor. Iron increases AKT phosphorylation that is prevented by DNTAK1 or DNp21ras. Iron causes ERK1/2 phosphorylation that is attenuated by DN-PI3K, prevented by DNp21ras, but unaffected by DNTAK1. Iron-induced TAK1 activity is not affected by the PI3K or MEK1 inhibitor, suggesting TAK1 is upstream of PI3K and MEK1. Iron increases interactions of TAK1 and PI3K with p21ras as demonstrated by co-immunoprecipitation and co-localization of these proteins with caveolin-1 as shown by immunofluorescent microscopy. Finally, filipin III, a caveolae inhibitor, abrogates iron-induced TAK1 and IKK activation. In conclusion, MEK1, TAK1, NF-kappa-inducing kinase, and PI3K are required for iron-induced IKK activation in hepatic macrophages and TAK1, PI3K, and p21ras physically interact in caveolae to initiate signal transduction.


Assuntos
Cavéolas/enzimologia , Quinase I-kappa B/metabolismo , Ferro/metabolismo , Fígado/enzimologia , MAP Quinase Quinase Quinases/metabolismo , Macrófagos/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Antibacterianos/farmacologia , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Filipina/farmacologia , Quinase I-kappa B/genética , Fígado/citologia , MAP Quinase Quinase Quinases/genética , Macrófagos/citologia , Masculino , Camundongos , Fosfatidilinositol 3-Quinases/genética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
17.
J Biol Chem ; 280(49): 40650-9, 2005 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-16216869

RESUMO

Depletion of peroxisome proliferator-activated receptor gamma (PPARgamma) represents one of the key molecular changes that underlie transdifferentiation (activation) of hepatic stellate cells in the genesis of liver fibrosis (Miyahara, T., Schrum, L., Rippe, R., Xiong, S., Yee, H. F., Jr., Motomura, K., Anania, F. A., Willson, T. M., and Tsukamoto, H. (2000) J. Biol. Chem. 275, 35715-35722; Hazra, S., Xiong, S., Wang, J., Rippe, R. A., Krishna, V., Chatterjee, K., and Tsukamoto, H. (2004) J. Biol. Chem. 279, 11392-11401). In support of this notion, ectopic expression of PPARgamma suppresses hepatic stellate cells activation markers, most notably expression of alpha1(I) procollagen. However, the mechanisms underlying this antifibrotic effect are largely unknown. The present study utilized deletion-reporter gene constructs of proximal 2.2-kb alpha1(I) procollagen promoter to demonstrate that a region proximal to -133 bp is where PPARgamma exerts its inhibitory effect. Within this region, two DNase footprints with Sp1 and reverse CCAAT box sites exist. NF-I, but not CCAAT DNA-binding factor/NF-Y, binds to the proximal CCAAT box in hepatic stellate cells. A mutation of this site almost completely abrogates the promoter activity. NF-I mildly but independently stimulates the promoter activity and synergistically promotes Sp1-induced activity. PPARgamma inhibits NF-I binding to the most proximal footprint (-97/-85 bp) and inhibits its transactivity. The former effect is mediated by the ability of PPARgamma to inhibit p300-facilitated NF-I binding to DNA as demonstrated by chromatin immunoprecipitation assay.


Assuntos
Colágeno Tipo I/genética , DNA/metabolismo , Proteína p300 Associada a E1A/fisiologia , Fígado/metabolismo , Fatores de Transcrição NFI/metabolismo , PPAR gama/fisiologia , Animais , Sequência de Bases , DNA/química , Pegada de DNA , Desoxirribonucleases , Ensaio de Desvio de Mobilidade Eletroforética , Deleção de Genes , Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Fígado/citologia , Masculino , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fatores de Transcrição NFI/antagonistas & inibidores , PPAR gama/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/análise , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão , Fator de Transcrição Sp1/metabolismo , Transfecção
18.
Hepatology ; 42(4): 905-14, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16175602

RESUMO

Nonalcoholic steatohepatitis is prevalent among obese individuals with excessive caloric intake, insulin resistance, and type II diabetes. However, no animal models exist that recapitulate this important association. This study produced and characterized steatohepatitis (SH) caused by intragastric overfeeding in mice. C57BL/6, tumor necrosis factor (TNF) type I receptor-deficient, and genetically matched wild type mice were fed via an implanted gastrostomy tube a high-fat diet for 9 weeks in the increasing amount up to 85% in excess of the standard intake. Animals were examined for weight gain, insulin sensitivity, and histology and biochemistry of liver and white adipose tissue (WAT). Overfed C57BL/6 mice progressively became obese, with 71% larger final body weights. They had increased visceral WAT, hyperglycemia, hyperinsulinemia, hyperleptinemia, glucose intolerance, and insulin resistance. Of these mice, 46% developed SH with increased plasma alanine aminotransferase (121 +/- 27 vs. 13 +/- 1 U/L), neutrophilic infiltration, and sinusoidal and pericellular fibrosis. Obese WAT showed increased TNFalpha and leptin expression and reciprocally reduced adiponectin expression. The expression of lipogenic transcription factors (SREBP-1c, PPARgamma, LXRalpha) was increased, whereas that of a lipolytic nuclear factor PPARalpha was reduced in SH. SH was associated with reduced cytochrome P450 (Cyp)2e1 but increased Cyp4a. TNF type I receptor deficiency did not prevent obesity and SH. In conclusion, forced overfeeding with a high-fat diet in mice induces obesity, insulin resistance, and SH in the absence of TNF signaling or Cyp2e1 induction.


Assuntos
Gorduras na Dieta/farmacologia , Ingestão de Alimentos , Fígado Gorduroso/etiologia , Gastrostomia , Cirrose Hepática/etiologia , Alanina Transaminase/sangue , Animais , Citocromo P-450 CYP2E1/metabolismo , Citocromo P-450 CYP4A/metabolismo , Modelos Animais de Doenças , Fígado Gorduroso/imunologia , Fígado Gorduroso/fisiopatologia , Intolerância à Glucose/etiologia , Intolerância à Glucose/imunologia , Intolerância à Glucose/fisiopatologia , Resistência à Insulina , Leptina/metabolismo , Cirrose Hepática/imunologia , Cirrose Hepática/fisiopatologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/patologia , Obesidade/etiologia , Obesidade/imunologia , Obesidade/fisiopatologia , PPAR alfa/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/metabolismo
19.
Hepatol Res ; 32(4): 202-12, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16107322

RESUMO

BACKGROUND AND AIMS: : A herbal prescription, Yan-gan-wan (YGW), has been known to offer hepatoprotective effects in Asian countries for years. This study investigated its mechanisms of action. METHODS: : The effects of YGW on CCl(4) induced liver damage were tested in mice and cultured hepatocytes. Microarray analysis screened genes affected by YGW. YGWs effects on the expression of cytochrome P450 (CYP) 2E1 and other isozymes were determined. YGWs effects on TNFalpha expression and NF-kappaB activation in Kupffer cells (KC), and TNFalpha promoter activity in RAW264.7 cells, were also assessed. RESULTS: : Administration of YGW reduced the plasma ALT, centrilobular necrosis, neutrophilic infiltration, and TNFalpha mRNA in the livers of mice acutely given CCl(4). The in vivo herb treatment reduced ALT release and necrosis of isolated hepatocytes directly exposed to CCl(4). Microarray analysis demonstrated marked reductions in CYP4A10 and 4A14 by YGW but no changes in other CYP isozymes as confirmed by immunoblot analysis. The herb treatment suppressed LPS-stimulated TNFalpha release in vivo and by cultured KC. Direct addition of the aqueous herb extract suppressed NF-kappaB activation by KC and TNFalpha promoter activity in RAW cells under LPS stimulation. This activity to suppress TNFalpha expression was largely separated into gel filtration fractions with the molecular size of 102-107Da. YGW also attenuated liver fibrosis induced by chronic treatment of CCl(4) or porcine serum. CONCLUSIONS: : The protective effects of YGW on CCl(4) hepatotoxicity are due in part to inhibition of KC NF-kappaB activation and TNFalpha expression by small water soluble molecules, and may also be related to suppressed hepatic expression of CYP4A10 and 4A14 that are considered as alternative prooxidant cytochromes.

20.
J Biol Chem ; 280(6): 4959-67, 2005 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-15537655

RESUMO

Hepatic stellate cells (HSC) undergo transdifferentiation (activation) from lipid-storing pericytes to myofibroblastic cells to participate in liver fibrogenesis. Our recent work demonstrates that depletion of peroxisome proliferator-activated receptor gamma (PPARgamma) constitutes one of the key molecular events for HSC activation and that ectopic expression of this nuclear receptor achieves the phenotypic reversal of activated HSC to the quiescent cells. The present study extends these findings to test a novel hypothesis that adipogenic transcriptional regulation is required for the maintenance of HSC quiescence. Comparative analysis of quiescent and activated HSC in culture reveals higher expression of putative adipogenic transcription factors such as CCAAT/enhancer-binding protein (C/EBP) alpha, C/EBPbeta, C/EBPdelta, PPARgamma, liver X receptor alpha, sterol regulatory element-binding protein 1c and of adipocyte-specific genes in the quiescent cells. Conversely, activated HSC have increased expression of PPARbeta, a transcription factor known to promote fatty acid oxidation. A treatment of activated HSC with the adipocyte differentiation mixture (isobutylmethylxanthine, dexamethasone, and insulin) or ectopic expression of PPARgamma or SREBP-1c in these cells, induces a panel of adipogenic transcription factors, reduces PPARbeta, and causes the phenotypic reversal to quiescent HSC. These results support the importance of adipogenic transcriptional regulation in HSC quiescence and provide a new framework for identifying novel molecular targets for the treatment of liver cirrhosis.


Assuntos
Adipócitos/metabolismo , Fígado/citologia , Fígado/metabolismo , Transcrição Gênica , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diferenciação Celular , Meios de Cultura/farmacologia , Primers do DNA/química , Proteínas de Ligação a DNA/metabolismo , Ácidos Graxos/metabolismo , Fibrose , Metabolismo dos Lipídeos , Fígado/patologia , Modelos Biológicos , Oxigênio/metabolismo , PPAR gama/metabolismo , Fenótipo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1 , Fatores de Tempo , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...