Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Immune Netw ; 17(4): 261-268, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28860955

RESUMO

Tumor necrosis factor-α (TNF-α) induces serum amyloid A (SAA) 3 among acute-phase proteins in mouse granulosa cells by activating NF-κB signaling via p55 TNF-α receptor type 1. However, the localization of SAA3 within the ovary is unknown. Here we investigated ovarian localization of SAA3 in a mouse ovulation model and in response to IL-1ß, a proinflammatory mediator. For the ovulation model, equine chorionic gonadotropin (eCG; 2.5 IU) was administered to mice subcutaneously (sc) to stimulate follicular development on day 25 of age and then 50 h after eCG, human chorionic gonadotropin (hCG; 2.5 IU) was administered sc to induce ovulation. The mouse ovulation model was characterized by the localization of CYP19 mRNA expression to granulosa layers of larger follicles. SAA3 mRNA, determined by in situ hybridization, was broadly expressed throughout the whole ovary. Granulosa layers and small follicles expressed higher SAA3 mRNA compared to thecal-interstitial layers and large follicles, respectively. Interestingly, atretic follicles contained cells expressing intense SAA3 mRNA. After ovulation, SAA3 mRNA expression was intensely evident in ruptured follicles and corpora lutea (CL). The intraperitoneal administration of IL-1ß revealed the intense and extensive appearance of specific cells expressing SAA3 mRNA around follicles and in CL. In addition, Gene Expression Omnibus (GEO) database analysis supported expression pattern of SAA3 mRNA observed in mouse ovulation model. Taken together, SAA3 was broadly distributed through the whole ovary, but intensely expressed in atretic follicles and CL. Furthermore, proinflammatory mediators could trigger the intense appearance of SAA3 around follicles and in CL.

2.
Endocrinology ; 151(7): 3407-19, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20444945

RESUMO

TNFalpha is an inflammatory-related cytokine that has inhibitory effects on gonadotropin- and cAMP-stimulated steroidogenesis and folliculogenesis. Because ovulation is an inflammatory reaction and TNF specifically induces serum amyloid A3 (SAA3) in mouse granulosa cells, the effect of cAMP on TNF-induced SAA3 promoter activity, mRNA and protein was investigated. Granulosa cells from immature mice were cultured with TNF and/or cAMP. TNF increased SAA3 promoter activity, mRNA, and protein, which were further increased by cAMP. cAMP alone increased SAA3 promoter activity, but SAA3 mRNA and protein remained undetectable. Thus, there appeared to be different mechanisms by which TNF and cAMP regulated SAA3 expression. SAA3 promoters lacking a nuclear factor (NF)-kappaB-like site or containing its mutant were not responsive to TNF but were responsive to cAMP. Among four CCAAT-enhancing binding protein (C/EBP) sites in the SAA3 promoter, the C/EBP site nearest the NF-kappaB-like site was required for TNF-induced SAA3. The C/EBP site at -75/-67 was necessary for responsiveness to cAMP. Dominant-negative C/EBP and cAMP response element-binding protein or short interfering RNA of C/EBPbeta blocked TNF- or cAMP-induced SAA3 promoter activity. The combination of TNF and cAMP increased C/EBPbeta protein above that induced by TNF or cAMP alone. Thus, cAMP in combination with TNF specifically induced C/EBPbeta protein, leading to enhanced SAA3 expression but requiring NF-kappaB in mouse granulose cells. In addition, like TNF, SAA inhibited cAMP-induced estradiol accumulation and CYP19 levels. These data indicate SAA may play a role in events occurring during the ovulation process.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , AMP Cíclico/farmacologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Isoformas de Proteínas/metabolismo , Proteína Amiloide A Sérica/genética , Fator de Necrose Tumoral alfa/farmacologia , Animais , Aromatase/genética , Northern Blotting , Western Blotting , Feminino , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Ovário/metabolismo , Regiões Promotoras Genéticas/genética , Receptores Tipo I de Fatores de Necrose Tumoral , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Chamariz do Fator de Necrose Tumoral
3.
J Cell Sci ; 123(Pt 6): 861-70, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20159967

RESUMO

Glycogen synthase kinase 3 beta (GSK-3beta) is constantly active in cells and its activity increases after serum deprivation, indicating that GSK-3beta might play a major role in cell survival under serum starvation. In this study, we attempted to determine how GSK-3beta promotes cell survival after serum depletion. Under full culture conditions (10% FBS), GSK-3beta inhibition with chemical inhibitors or siRNAs failed to induce cell death in human prostate cancer cells. By contrast, under conditions of serum starvation, a profound necrotic cell death was observed as evidenced by cellular morphologic features and biochemical markers. Further analysis revealed that GSK-3beta-inhibition-induced cell death was in parallel with an extensive autophagic response. Interestingly, blocking the autophagic response switched GSK-3beta-inhibition-induced necrosis to apoptotic cell death. Finally, GSK-3beta inhibition resulted in a remarkable elevation of Bif-1 protein levels, and silencing Bif-1 expression abrogated GSK-3beta-inhibition-induced autophagic response and cell death. Taken together, our study suggests that GSK-3beta promotes cell survival by modulating Bif-1-dependent autophagic response and cell death.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose , Autofagia , Quinase 3 da Glicogênio Sintase/metabolismo , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultura Livres de Soro , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Humanos , Masculino , Modelos Biológicos , Necrose , Inibidores de Proteínas Quinases/farmacologia
4.
Int J Cancer ; 126(3): 764-74, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19642108

RESUMO

The androgen receptor (AR) is the most critical factor in prostate cancer progression. We previously demonstrated that silencing the AR using 2 unique small interfering RNAs (no. 8 and no. 31 AR siRNA) induces apoptotic cell death in AR-positive prostate cancer cells. To develop this AR siRNA technique into a therapy for prostate cancers, we generated an adeno-associated virus (AAV) vector to stably express a short hairpin-structured RNA (shRNA) against the AR gene in vivo. In addition to the no. 8 AR shRNA (ARHP8), we also screened a group of AR shRNAs with different sequences and identified a less effective AR shRNA (ARHP4) that was used as an shRNA control. An empty AAV vector (AAV-GFP) was used as a negative control. Intratumoral injection of AAV-ARHP8 viruses significantly suppressed tumor growth of xenografts derived from either androgen-responsive or castration-resistant prostate cancer cells. Most interestingly, systemic delivery of the AAV-ARHP8 but not AAV-ARHP4 or AAV-GFP viruses via tail vein injection eliminated xenografts within 10 days. Further analysis revealed that AAV-ARHP8 viruses dramatically reduced the expression of AR-regulated cellular survival genes and caused a dramatic apoptotic response. Taken together, our data strongly suggest that AAV-ARHP8 viruses induced a strong AR gene silencing in vivo and that systemic delivery of ARHP8 siRNA via an AAV vector or any other means might be considered as novel gene therapy for prostate cancers.


Assuntos
Adenocarcinoma/terapia , Dependovirus/genética , Inativação Gênica , Vetores Genéticos/uso terapêutico , Neoplasias da Próstata/terapia , RNA Interferente Pequeno/uso terapêutico , Receptores Androgênicos/genética , Adenocarcinoma/cirurgia , Antagonistas de Receptores de Andrógenos , Animais , Apoptose , Linhagem Celular Tumoral/transplante , Terapia Combinada , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Injeções Intralesionais , Masculino , Camundongos , Camundongos Nus , Orquiectomia , Neoplasias da Próstata/cirurgia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Mol Cell Biochem ; 318(1-2): 191-200, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18642057

RESUMO

In order to evaluate the role of Src tyrosine kinase in thecal cell steroidogenesis, a pharmacological approach was utilized by treating enriched populations of mouse ovarian theca-interstitial cells in vitro with a direct Src kinase inhibitor, PP2. Inhibition of Src with PP2 increased both basal and forskolin-stimulated androstenedione secretion, and increased cytochrome P450 17-alpha hydroxylase-lyase (CYP17) promoter activity and steady state mRNA. PP2 did not change thecal levels of StAR mRNA. Inhibition of mitogen-activated protein kinase kinase, a downstream regulator of Src activity, using PD98059 also increased forskolin-stimulated secretion of androstenedione above forskolin alone, but had no effect on basal secretion of androstenedione. Src inhibition increased mitogen-activated protein kinase phosphatase-1 protein and decreased phosphorylation of SF-1, which correlated with increased CYP17 promoter activity and mRNA levels. These results implicate Src tyrosine kinase in the regulation of CYP17 and thecal androgen secretion.


Assuntos
Androstenodiona/metabolismo , Esteroide 17-alfa-Hidroxilase/metabolismo , Células Tecais/enzimologia , Células Tecais/metabolismo , Quinases da Família src/metabolismo , Animais , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Colforsina/farmacologia , AMP Cíclico/metabolismo , Fosfatase 1 de Especificidade Dupla/genética , Fosfatase 1 de Especificidade Dupla/metabolismo , Feminino , Flavonoides/farmacologia , MAP Quinase Quinase 1/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas , Pirimidinas/farmacologia , Esteroide 17-alfa-Hidroxilase/genética , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Células Tecais/efeitos dos fármacos , Quinases da Família src/antagonistas & inibidores
7.
Prostate ; 68(4): 453-61, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18196538

RESUMO

BACKGROUND: Recently we reported that silencing the androgen receptor (AR) gene reduced Bcl-xL expression that was associated with a profound apoptotic cell death in prostate cancer cells. In this study we further investigated AR-regulated Bcl-xL expression. METHODS: Prostate cancer cell line LNCaP and its sublines, LNCaP/PURO and LNCaP/Bclxl, were used for cell proliferation assay and xenograft experiments in nude mice. Luciferase gene reporters driven by mouse or human bcl-x gene promoter were used to determine androgen regulation of Bcl-xL expression. RT-PCR and Western blot assays were conducted to assess Bcl-xL gene expression. Chromatin immunoprecipitation assay was performed to determine AR interaction with Bcl-xL promoter. Bcl-xL-induced alteration of gene expression was examined using cDNA microarray assay. RESULTS: In cultured prostate cancer LNCaP cells, androgen treatment significantly increased Bcl-xL expression at mRNA and protein levels via an AR-dependent mechanism. Promoter analyses demonstrated that the AR mediated androgen-stimulated bcl-x promoter activation and that the AR interacted with bcl-x promoter. Enforced expression of Bcl-xL gene dramatically increased cell proliferation in vitro and promoted xenograft tumor growth in vivo. Genome-wide gene profiling analysis revealed that Bcl-xL expression was significantly higher in metastatic and castration-resistant diseases compared to normal prostate tissues or primary cancers. Bcl-xL overexpression significantly increased the expression of cyclin D2, which might be responsible for Bcl-xL-induced cell proliferation and tumor growth. CONCLUSIONS: Taken together, our data strongly suggest that androgen stimulates Bcl-xL expression via the AR and that increased Bcl-xL expression plays a versatile role in castration-resistant progression of prostate cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/patologia , Neoplasias da Próstata/fisiopatologia , Receptores Androgênicos/metabolismo , Proteína bcl-X/genética , Androgênios/metabolismo , Animais , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Ciclina D2 , Ciclinas/metabolismo , Progressão da Doença , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Orquiectomia , Regiões Promotoras Genéticas/fisiologia , Neoplasias da Próstata/genética
8.
Toxicol Appl Pharmacol ; 223(1): 66-72, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17594909

RESUMO

The aryl hydrocarbon receptor (AHR) mediates the toxicity of a variety of environmental chemicals. Although little is known about the physiological role of the AHR, studies suggest that it plays an important role in regulating ovulation because Ahr deficient (AhRKO) mice have a reduced number of ovulations compared to wild-type (WT) mice. The reasons for the reduced ability of AhRKO mice to ovulate are unknown. Normal ovulation, however, requires estrous cyclicity, appropriate luteinizing hormone (LH) and follicle-stimulating hormone (FSH) levels, and LH and FSH responsiveness. Thus, the purpose of this study was to test the hypothesis that Ahr deletion regulates ovulation by altering cyclicity, FSH and LH levels, follicle-stimulating hormone receptor (Fshr) and luteinizing hormone receptor (Lhcgr) levels and/or gonadotropin responsiveness. The data indicate that AhRKO and WT mice have similar levels of FSH and LH, but AhRKO mice have reduced Fshr and Lhcgr mRNA levels compared to WT mice. Furthermore, AhRKO ovaries contain fewer corpora lutea compared to WT ovaries after 5 IU equine chorionic gonadotropin (eCG) treatment. Lastly, both AhRKO and WT mice ovulate a similar number of eggs in response to 5 IU human chorionic gonadotropin (hCG), but AhRKO mice ovulate fewer eggs than WT mice in response to 2.5 IU and 1.25 IU hCG. Collectively, these data indicate that AhRKO follicles have a reduced capacity to ovulate compared to WT follicles and that this is due to reduced responsiveness to gonadotropins. Thus, in addition to mediating toxicity of environmental chemicals, the Ahr is required for normal ovulation.


Assuntos
Gonadotropina Coriônica/farmacologia , Estro/sangue , Hormônio Foliculoestimulante/sangue , Hormônio Luteinizante/sangue , Ovário/metabolismo , Receptores de Hidrocarboneto Arílico/deficiência , Animais , Corpo Lúteo/efeitos dos fármacos , Corpo Lúteo/patologia , Estro/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Inativação Gênica , Masculino , Camundongos , Camundongos Knockout , Ovário/efeitos dos fármacos , Ovário/patologia , Ovulação/efeitos dos fármacos , Inibição da Ovulação/genética , RNA Mensageiro/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores do FSH/genética , Receptores do FSH/metabolismo , Receptores do LH/genética , Receptores do LH/metabolismo
9.
Prostate ; 67(9): 976-88, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17440966

RESUMO

BACKGROUND: Lithium is an existing drug for bipolar disorder and its uptake was recently linked to reduced tumor incidence compared to the general population. The major target of lithium action is glycogen synthase kinase 3 (GSK-3). Since GSK-3 expression and activation are associated with prostate cancer progression, the anti-cancer potential of lithium on prostate cancer was investigated in this study. METHODS: Multiple prostate cancer cell lines were treated with lithium chloride (LiCl). Cell proliferation and cell cycle distribution were analysed. DNA replication was determined using BrdU labeling assay. Genome-wide screening of gene expression was performed using cDNA microarray assay. GSK-3beta gene-specific silencing was conducted using small interferencing RNA (siRNA) transfection. E2 factor (E2F) transactivation was evaluated using reporter gene assay and E2F-DNA interaction was determined with chromatin-immunoprecipitation assay (ChIP). RESULTS: LiCl significantly inhibited cell proliferation, which was associated with reduced DNA replication and S-phase cell cycle arrest. LiCl significantly decreased the expression of multiple DNA replication-related genes, including cell division cycle 6 (cdc6), cyclin A, cyclin E, and cdc25C, which are regulated by E2F factor during cell cycle. A novel GSK-3-specific inhibitor TDZD-8 and GSK-3beta siRNA also suppressed the expression of these E2F target genes, indicating that LiCl-induced anti-cancer effect was associated with GSK-3beta inhibition. Furthermore, LiCl suppressed E2F transactivation by interrupting the interaction of E2F1 factor with its target gene promoter. CONCLUSIONS: These data indicated that LiCl suppresses cancer cell proliferation by disrupting E2F-DNA interaction and subsequent E2F-mediated gene expression in prostate cancer.


Assuntos
Divisão Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Lítio/farmacologia , Fase S/genética , Linhagem Celular Tumoral , Primers do DNA , Humanos , Masculino , Proteínas de Neoplasias/genética , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fase S/efeitos dos fármacos
10.
Carcinogenesis ; 28(3): 572-83, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17032658

RESUMO

Aggressive androgen-independent (also termed as hormone-refractory) prostate cancer is a major clinical obstacle because there is no means to cure. Previous studies have shown that Akt activation is associated with prostate cancer progression from androgen-dependent to androgen-independent stage. However, its causative role in this process has not been established. One of the major limitations is the lack of a well-controlled inducible system to study Akt involvement. Recently, we developed a novel inducible Akt (iAKT) system based on a chemically induced dimerization (CID) approach. This system allows for conditional activation of Akt in a physiological setting. Utilizing this iAKT system, we found that Akt activation prevented cell death after serum withdrawal and promoted cell proliferation in the absence of androgen in vitro in human prostate cancer LNCaP cells, which should stop growing after androgen withdrawal or even die after serum starvation. The iAKT-induced death protection and growth promotion were further demonstrated in vivo using a transgenic mouse model that expresses the iAKT system conditionally in the prostate epithelium. Most importantly, in a mouse xenograft model derived from LNCaP cells, iAKT activation promoted tumor growth in castrated animals by enhancing cell proliferation and inhibiting apoptosis. Taken together, our data suggest that Akt activation is playing a causative role in androgen-independent progression of prostate cancer. This study provides a significant relevance of Akt-targeted therapy for hormone-refractory prostate cancers.


Assuntos
Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Androgênios/fisiologia , Animais , Divisão Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Meios de Cultura , Dimerização , Progressão da Doença , Ativação Enzimática , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/enzimologia , Transplante Heterólogo
11.
Biol Reprod ; 76(2): 224-31, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17065602

RESUMO

It is believed that a finite pool of primordial follicles is established during embryonic and neonatal life. At birth, the mouse ovary consists of clusters of interconnected oocytes surrounded by pregranulosa cells. Shortly after birth these structures, termed germ cell cysts or nests (GCN), break down to facilitate primordial follicle formation. Tumor necrosis factor alpha (TNF) is a widely expressed protein with myriad functions. TNF is expressed in the ovary and may regulate GCN breakdown in rats. We investigated whether it participates in GCN breakdown and follicle formation in mice by using an in vitro ovary culture system as well as mutant animal models. We found that TNF and both receptors (TNFRSF1A and TNFRSF1B) are expressed in neonatal mouse ovaries and that TNF promotes oocyte death in neonatal ovaries in vitro. However, deletion of either receptor did not affect follicle endowment, suggesting that TNF does not regulate GCN breakdown in vivo. Tnfrsf1b deletion led to an apparent acceleration of follicular growth and a concomitant expansion of the primordial follicle population. This expansion of the primordial follicle population does not appear to be due to decreased primordial follicle atresia, although this cannot be ruled out completely. This study demonstrates that mouse oocytes express both TNF receptors and are sensitive to TNF-induced death. Additionally, TNFRSF1B is demonstrated to be an important mediator of TNF function in the mouse ovary and an important regulator of folliculogenesis.


Assuntos
Camundongos/metabolismo , Folículo Ovariano/crescimento & desenvolvimento , Ovário/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Animais Recém-Nascidos , Morte Celular/fisiologia , Feminino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oócitos/metabolismo , Oócitos/fisiologia , Técnicas de Cultura de Órgãos , Ovário/citologia , Isoformas de Proteínas/metabolismo
12.
Endocrine ; 27(1): 17-24, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16077166

RESUMO

TNF is known to suppress gonadotropin-induced steroid secretion by Leydig cells. However, the mechanisms by which this occurs are largely unknown. Because expression of many steroidogenic proteins is regulated by the PKA pathway, effects of TNF on CRE activity were examined using MA-10 mouse Leydig tumor cells. The cells were transfected with a CRE-luciferase construct, and stimulated with either LH or 8Br-cAMP in the presence or absence of TNF. TNF suppressed, LH-stimulated and 8Br-cAMP stimulated CRE activity. TNF also suppressed CRE activity stimulated with a PKA expression vector. Further experiments suggested that the effect of TNF on CRE activity was not mediated by the NF-kappaB pathway. TNF did not affect levels of either CREB or phospho-CREB in whole cell lysates; however, TNF decreased both CREB and phospho-CREB in nuclear extracts in a time-dependent manner. The decrease in nuclear CREB is likely to be a major mechanism of the suppressive effects of TNF on steroidogenesis in MA-10 Leydig cells.


Assuntos
Núcleo Celular/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/antagonistas & inibidores , Tumor de Células de Leydig/metabolismo , Neoplasias Testiculares/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Western Blotting , Proteína de Ligação a CREB , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Genes Reporter , Luciferases/genética , Masculino , Camundongos , NF-kappa B/genética , NF-kappa B/fisiologia , Proteínas Nucleares/metabolismo , Radioimunoensaio , Esteroides/biossíntese , Transativadores/metabolismo , Transfecção
13.
Endocrine ; 26(2): 169-76, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15888929

RESUMO

The role of Src tyrosine kinase in regulating reproductive processes in female mice was investigated using Src wild-type (+/+), heterozygous (+/-), and knockout (-/-) mice. Ovarian Src kinase activity transiently increased in Src +/+ mice following eCG administration. Src knockout mice were infertile. Estrous cycles and vaginal opening in Src knockouts were variable and altered compared with Src +/+ and +/- mice. Follicle development was compromised in Src knockout mice as evidenced by reduced numbers of large pre-antral and antral follicles compared to Src +/+ mice. Corpora lutea were not observed in the ovaries of Src knockout mice; however, administration of eCG and hCG did result in ovulation. Serum LH and FSH on d 40 and 52 of age did not differ between Src wild-type and knockout females. Results from these studies reveal that female Src knockout mice are infertile due to reduced follicle development and anovulation.


Assuntos
Folículo Ovariano/enzimologia , Reprodução/fisiologia , Quinases da Família src/deficiência , Animais , Anovulação/fisiopatologia , Gonadotropina Coriônica/fisiologia , Ciclo Estral/fisiologia , Feminino , Hormônio Foliculoestimulante/sangue , Histocitoquímica , Hormônio Luteinizante/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , RNA Mensageiro/química , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Quinases da Família src/genética , Quinases da Família src/fisiologia
14.
Toxicol Sci ; 85(1): 560-71, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15703265

RESUMO

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) affects glycemia due to reduced gluconeogenesis; when combined with a reduction in feed intake, this culminates in decreased body weight. We investigated the effects of steady-state levels of TCDD (loading dose rates of 0.0125, 0.05, 0.2, 0.8, and 3.2 microg/kg) or approximately isoeffective dose rates of 1,2,3,4,7,8-hexachlorodibenzo-p-dioxin (HxCDD) (loading dose rates of 0.3125, 1.25, 5, 20, and 80 microg/kg) on body weight, phosphoenolpyruvate carboxykinase (PEPCK) mRNA expression and activity, and circulating concentrations of insulin, glucose, and insulin-like growth factor-I (IGF-I), and expression of hepatic phosphorylated AMP kinase-alpha (p-AMPK) protein in female Sprague-Dawley rats (approximately 250 gm) at 2, 4, 8, 16, 32, 64, and 128 days after commencement of treatment. At the 0.05 and 1.25 microg/kg loading dose rates of TCDD and HxCDD, respectively, there was a slight increase in body weight as compared to controls, whereas at the 3.2 and 80 microg/kg loading dose rates of TCDD and HxCDD, respectively, body weight of the rats was significantly decreased. TCDD and HxCDD also inhibited PEPCK activity in a dose-dependent fashion, as demonstrated by reductions in PEPCK mRNA and protein. Serum IGF-I levels of rats treated initially with 3.2 microg/kg TCDD or 80 microg/kg HxCDD started to decline at day 4 and decreased to about 40% of levels seen in controls after day 16, remaining low for the duration of the study. Eight days after initial dosing, hepatic p-AMPK protein was increased in a dose-dependent manner with higher doses of TCDD and HxCDD. There was no effect with any dose of TCDD or HxCDD on circulating insulin or glucose levels. In conclusion, doses of TCDD or HxCDD that began to inhibit body weight in female rats also started to inhibit PEPCK, inhibited IGF-I, while at the same time inducing p-AMPK.


Assuntos
Peso Corporal/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Fator de Crescimento Insulin-Like I/metabolismo , Fígado/efeitos dos fármacos , Dibenzodioxinas Policloradas/análogos & derivados , Dibenzodioxinas Policloradas/toxicidade , Transdução de Sinais/efeitos dos fármacos , Adenilato Quinase/biossíntese , Administração Oral , Animais , Relação Dose-Resposta a Droga , Feminino , Fígado/enzimologia , Fígado/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
15.
Endocrinology ; 145(5): 2245-52, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14749357

RESUMO

TNF-alpha has significant inhibitory effects on steroidogenesis and folliculogenesis and is associated with several inflammatory responses. Because ovulation is an inflammatory reaction, the effects of TNF on the family of acute-phase proteins in granulosa cells were investigated. Granulosa cells from immature mice at 28 d of age were cultured in the presence of 10 ng TNF/ml for 24 h. Serum amyloid A3 (SAA3), a main acute-phase protein, was induced by TNF in granulosa cells. The other isoforms of serum amyloid proteins SAA1, SAA2, and SAA4 were neither expressed in granulosa cells nor induced by TNF. TNF did not induce SAA3 mRNA in granulosa cells from TNF receptor type 1 (TNFR1) knockout mice, although SAA3 mRNA was induced within 3 h after TNF treatment in wild-type cells. Two SAA3 promoters, -617/+73 and -198/+73, were responsive to TNF and to p65, a component of the TNF signaling molecule nuclear factor (NF)-kappaB. The -106/+73 promoter of SAA3 lacking a NF-kappaB-like site was not responsive to TNF or p65. In granulosa cells from TNFR1 knockout mice, the SAA3 promoter (-198/+73) was responsive to transfection with the p65 component of NF-kappaB, but neither TNF treatment nor overexpression of the p50 component of NF-kappaB increased promoter activity. Similar results were observed in the murine ovarian granulosa tumor cell line (OV3121-1). Overexpression of the inhibitor of NF-kappaB (called IkappaB) blocked SAA3 promoter activity induced by TNF and by p65 in OV3121-1 cells. Closer analysis of deletion mutants of the SAA3 promoter revealed the necessity of a NF-kappaB like site for responsiveness to TNF in the OV3121-1 cells. TNF rapidly increased p65 in OV3121-1 nuclei when compared with controls not treated with TNF. TNF also increased phospho-IkB and SAA3 in whole-cell homogenates as determined by Western blots. Thus, TNF likely increased SAA3 promoter activity and protein by activating NF-kappaB signaling via TNFR1 in mouse granulosa cells. SAA3 is a novel gene in granulosa cells with yet unknown functions in the ovary.


Assuntos
Células da Granulosa/metabolismo , Proteína Amiloide A Sérica/biossíntese , Fator de Necrose Tumoral alfa/fisiologia , Animais , Antígenos CD/fisiologia , Sítios de Ligação , Western Blotting , Células Cultivadas , Feminino , Deleção de Genes , Expressão Gênica , Células da Granulosa/efeitos dos fármacos , Proteínas I-kappa B/genética , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese , NF-kappa B/genética , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas/genética , Isoformas de Proteínas/biossíntese , RNA Mensageiro/análise , Receptores do Fator de Necrose Tumoral/deficiência , Receptores do Fator de Necrose Tumoral/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Amiloide A Sérica/genética , Fator de Necrose Tumoral alfa/farmacologia
16.
Endocrinology ; 145(3): 1218-26, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14617571

RESUMO

TNF alpha has significant in vitro effects on steroidogenesis and folliculogenesis and reproductive alterations occur in TNF receptor type 1 (TNFR1) knockout mice. The present study investigated the effect of in vitro TNF on granulosa cell proliferation from immature mice at 28 d of age, with emphasis on intracellular signaling that regulates granulosa cell proliferation. TNF dose dependently increased granulosa cell proliferation and the proto-oncogene c-Jun protein. However, other Jun family members such as JunD was expressed constitutively and JunB was not expressed. In vitro TNF did not increase c-Jun and proliferation in granulosa cells from TNFR1 knockout mice. The time course of TNF-induced c-Jun revealed biphasic patterns of short-term (3 h) and long-term (24 h) induction. The time courses of Ser63- and Ser73-phospho c-Jun coincided with changes in total c-Jun. Among MAPK cascades, stress-activated protein kinase/c-Jun-NH(2)-teminal kinase signaling was increased transiently in TNF-treated cells, whereas p38MAPK and ERK1 and 2 were not changed. In addition, overexpression of nuclear factor-kappa B and addition of ceramide and 8-bromo-cAMP did not increase c-Jun or proliferation. Antisense oligonucleotides for c-Jun blocked cell proliferation induced by TNF. In conclusion, the above results demonstrate that TNF increased c-Jun by activating stress-activated protein kinase/c-Jun-NH(2)-teminal kinase signaling via TNFR1 in mouse granulosa cells, and the induced c-Jun resulted in increased cell proliferation.


Assuntos
Antígenos CD/metabolismo , Antineoplásicos/farmacologia , Células da Granulosa/citologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fatores Etários , Animais , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Receptores Tipo I de Fatores de Necrose Tumoral , Fator de Transcrição AP-1/metabolismo
17.
Endocrine ; 25(2): 147-54, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15711029

RESUMO

Src tyrosine kinase belongs to a non-receptor tyrosine kinase family and has been shown to be involved in G protein-coupled receptor desensitization and internalization. Stimulation of ovarian thecal cells with lutein-izing hormone (LH) activates adenylyl cyclase via a G protein-coupled LH receptor leading to an increase in cAMP. Subsequently, cAMP activates protein kinase A (PKA) that increases steroidogenesis. In order to evaluate the role of Src in thecal cell steroidogenesis, a pharmacological approach was utilized by treating a population of mouse ovarian theca-enriched cells (TEC) in vitro with two Src inhibitors, geldanamycin (GA) and herbimycin A (HA). Treatment of TEC with either GA or HA increased basal androstenedione secretion without alteration of cAMP. In the presence of forskolin, GA and HA treatment further increased androstenedione secretion. RT-PCR analysis of RNA from cells treated with GA for 8, 24, and 48 h revealed that GA increased cytochrome P450 17alpha-hydroxylase/lyase (CYP17) mRNA at 48 h. CYP17 promoter activity also increased after treatment of cells with GA and after co-transfection with a Src dominant negative plasmid. Inhibition of PKA using H89 blocked the effect GA and HA on androstenedione secretion. These results indicate that the pharmacological inhibitors of Src, GA and HA, tested in vitro increased thecal CYP17 promoter activity, CYP17 mRNA, and androstenedione secretion. In addition, GA and HA induced thecal androstenedione secretion may be cAMP independent but possibly requires PKA.


Assuntos
Androstenodiona/metabolismo , Ovário/fisiologia , Esteroide 17-alfa-Hidroxilase/biossíntese , Quinases da Família src/metabolismo , 3-Hidroxiesteroide Desidrogenases/genética , 3-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Benzoquinonas , Colforsina/farmacologia , AMP Cíclico/metabolismo , Feminino , Isoquinolinas/farmacologia , Lactamas Macrocíclicas , Camundongos , Camundongos Endogâmicos C57BL , Ovário/enzimologia , Ovário/metabolismo , Progesterona/metabolismo , Regiões Promotoras Genéticas , Inibidores de Proteínas Quinases/farmacologia , Quinonas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rifabutina/análogos & derivados , Esteroide 17-alfa-Hidroxilase/genética , Sulfonamidas/farmacologia , Células Tecais/enzimologia , Células Tecais/metabolismo , Células Tecais/fisiologia , Quinases da Família src/antagonistas & inibidores
18.
Toxicology ; 195(1): 1-17, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14698564

RESUMO

Interactions between the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and protein kinase C (PKC) signaling pathways are governed in cell and tissue-specific manners, albeit the physiological significance of which is unclear. This research sought to define the effects of TCDD on the PKC pathway using a mouse ovarian surface epithelial cancer cell line (ID8). Phorbol-12-myristate-13-acetate (PMA) potentiated (1 nM) TCDD-induced 7-ethoxyresorufin-O-deethylase (EROD) activity after 24h of treatment, and pre-treatment with (1 microM) of either a general PKC inhibitor (BisI) or PKCdelta-specific inhibitor (Rotterlin) abolished the potentiation indicating that activation of PKC enhances TCDD signal transduction. Western blot analysis revealed that unstimulated ID8 cells express PKCalpha, beta, epsilon, tau, lambda and RACK1. PKCgamma, eta, theta and DGKtheta were not detected. TCDD (1 nM) increased PKCdelta protein approximately eight-fold after 24h of treatment and this effect was dose-dependent (0.1-100 nM); other PKC isoforms and related signaling proteins tested were unaffected by TCDD treatment. Immunofluorescent microscopy revealed that TCDD (1 nM) promoted the subcellular redistribution of PKCdelta, from the cytoplasm and the nucleus to the perinuclear area after 2h of treatment, however, after 24h of treatment PKCdelta was observed in nuclear structures that resembled nucleoli. TCDD (1 nM) also increased total PKC and PKCdelta-specific kinase activities in biphasic time-responsive manners. Total PKC and PKCdelta-specific activities increased after 1-2h of treatment. Then TCDD increased the total PKC activity again after 12h of treatment, whereas, PKCdelta-specific activity resurged at 24h and remained elevated at 48 h after treatment. The results indicate that TCDD preferentially induces PKCdelta protein expression and phosphotransferase activity, and its membrane translocation, indicating a potential intracellular role for PKCdelta as an effector molecule for TCDD-mediated biological events in this ovarian cancer cell line.


Assuntos
Poluentes Ambientais/toxicidade , Células Epiteliais/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Proteína Quinase C/metabolismo , Animais , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Clonagem Molecular , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células Epiteliais/enzimologia , Feminino , Imuno-Histoquímica , Isoenzimas/antagonistas & inibidores , Isoenzimas/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/patologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-delta , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Células Tumorais Cultivadas
19.
Biochem Biophys Res Commun ; 309(2): 377-83, 2003 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-12951060

RESUMO

The vast majority of ovarian cancers originate in the ovarian surface epithelium. Unfortunately, there is a lack of appropriate animal models for ovarian cancer research. Spontaneously transformed mouse ovarian surface epithelial cells may provide a faithful animal model for human ovarian cancer. One such cell line (ID8) has been partially characterized. ID8 cells demonstrate constitutive Src tyrosine kinase activation with resulting phosphatidylinositol-3-kinase activation and Akt and forkhead phosphorylation. In addition, focal adhesion kinase is constitutively phosphorylated at tyrosine 925, a Src phosphorylation site, resulting in increased Ras activation. These features are common to human ovarian cancer cell lines. Inhibition of Src enhances the cell killing effects of both paclitaxel and cisplatinum. Finally, Src inhibition restores sensitivity of a drug resistant ID8 cell line. The ID8 mouse ovarian cancer cell line presents new opportunities to study ovarian cancer progression and pre-therapeutic trials in an immune competent background.


Assuntos
Cisplatino/farmacologia , Neoplasias Ovarianas/metabolismo , Paclitaxel/farmacologia , Proteínas Serina-Treonina Quinases , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Feminino , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia
20.
Toxicol Appl Pharmacol ; 183(3): 179-88, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12383709

RESUMO

Immature Sprague-Dawley rats received daily doses of indole-3-carbinol (I3C, 0-1.5 g/kg/day), 3,3'-diindolymethane (DIM, 0-400 mg/kg/day), tamoxifen (TAM, 0-0.5 mg/kg/day), or vehicle to determine if their antiestrogenic effects occur by the same mechanism and whether I3C's action is mediated by DIM. Follicular development was induced on day 24 of age by equine chorionic gonadotropin (eCG, 5 IU) 1 day after the initial dose. In a hormone replacement study, human chorionic gonadotropin (hCG, 10 IU sc, 48 h post-eCG) was used to mimic a normal preovulatoy luteinizing hormone (LH) surge following treatment with either I3C or TAM. Blood and ovaries were collected throughout follicular development and the number of ova shed was measured on the morning following expected ovulation (72 h post-eCG). I3C but not TAM reduced body weight gain at higher doses after 4 days of dosing. Ovarian weight gain and ovulation were inhibited by both I3C and TAM in a dose-dependent fashion. During the preovulatory period, both I3C and TAM blocked normal LH and follicle-stimulating hormone (FSH) surges and suppressed serum progesterone (P(4)) profoundly without changing circulating levels of estrogen (E(2)). At the time of expected ovulation, serum E(2) was increased in rats receiving I3C or tamoxifen, whereas serum P(4) was dose-dependently decreased. DIM exerted no significant effects on any of the endpoints studied, even at the highest dose, indicating that the antiestrogenic effects of I3C are not mediated by this metabolite of I3C. hCG successfully restored ovarian weight gain and ovulation in TAM-treated rats. However, hCG only partially reversed the blockage of ovulation by I3C, although ovarian weight gain was restored to normal. In summary, both I3C and TAM block ovulation by altering preovulatory concentrations of LH and FSH, but I3C appears to exert its effect(s) by (a) different mechanism(s) of action. I3C seems to act at both the ovarian and hypothalamic levels by mechanisms similar to those seen in TCDD-treated rats, whereas TAM appears to act only on the hypothalamic-pituitary axis as an anti-estrogen.


Assuntos
Indóis/farmacologia , Ovulação/efeitos dos fármacos , Tamoxifeno/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Gonadotropinas/sangue , Tamanho do Órgão/efeitos dos fármacos , Ovário/citologia , Ovário/efeitos dos fármacos , Ovário/crescimento & desenvolvimento , Ratos , Ratos Sprague-Dawley , Esteroides/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA