Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Br J Nutr ; 113(9): 1384-95, 2015 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-25850566

RESUMO

The objective of the present study was to find the optimum dose of flaxseed that would decrease PG and alter oestrogen pathway endpoints implicated in ovarian cancer. In the study, four groups of fifty 1.5-year-old chickens were fed different amounts of flaxseed (0, 5, 10 or 15% of their total diet) for 4 months and were then killed to collect blood and tissues. Levels of flaxseed lignan metabolites, Enterolactone (EL) and Enterodiol (ED) were measured in the serum, liver and ovaries by liquid chromatography-MS/MS, and n-3 and n-6 fatty acid (FA) levels were measured by GC. The effects of the varied flaxseed doses were assessed by measuring levels of PGE2 and oestrogen metabolites (16-hydroxyestrone (16-OHE1) and 2-hydroxyestrone (2-OHE1)) as well as by analysing the expression of the oestradiol metabolising enzymes CYP3A4 (cytochrome p450, family 3, subfamily A, polypeptide 4), CYP1B1 (cytochrome p450, family 1, subfamily B, polypeptide 1) and CYP1A1 (cytochrome p450, family 1, subfamily A, polypeptide 1) and that of oestrogen receptor α (ERα) in the ovaries. The ratio of n-3:n-FA increased with an increase in flaxseed supplementation and corresponded to a dose-dependent decrease in cyclo-oxygenase-2 protein and PGE2 levels. EL and ED increased in the serum, liver and ovaries with increased concentrations of flaxseed. Flaxseed decreased the expression of ERα in the ovaries. The ratio of 2-OHE1:16-OHE1 in the serum increased significantly in the 15% flaxseed diet, and there was a corresponding increase in CYP1A1 in the liver and decrease in CYP3A4 in the ovaries. CYP1B1 mRNA also decreased with flaxseed diet in the ovaries. The 15% flaxseed-supplemented diet significantly decreased inflammatory PGE2, ERα, CYP3A4, CYP1B1 and 16-OHE1, but it increased CYP1A1 and 2-OHE1, which thus reduced the inflammatory and pro-carcinogenic micro-environment of the ovaries.


Assuntos
Anticarcinógenos/administração & dosagem , Galinhas , Dieta/veterinária , Linho , Neoplasias Ovarianas/prevenção & controle , Ovário/metabolismo , 4-Butirolactona/análogos & derivados , 4-Butirolactona/análise , 4-Butirolactona/sangue , Animais , Ciclo-Oxigenase 1/análise , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/análise , Ciclo-Oxigenase 2/genética , Citocromo P-450 CYP1A1/análise , Citocromo P-450 CYP1B1/análise , Citocromo P-450 CYP3A/análise , Suplementos Nutricionais , Dinoprostona/análise , Receptor alfa de Estrogênio/análise , Estrogênios/metabolismo , Ácidos Graxos Ômega-3/análise , Ácidos Graxos Ômega-6/análise , Feminino , Hidroxiestronas/análise , Lignanas/análise , Lignanas/sangue , Lignanas/metabolismo , Fígado/química , Ovário/química , RNA Mensageiro/análise
2.
Gynecol Oncol ; 130(3): 620-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23707669

RESUMO

OBJECTIVE: Ovarian cancer is the most lethal gynecological malignancy. Prevention may be the best approach to reduce ovarian cancer. Flaxseed is the richest vegetable source of omega-3 fatty acids which may be effective in the prevention of ovarian cancer. Prostaglandin E2 (PGE2) is the most pro-inflammatory ecoisanoid and one of the downstream products of two isoforms of cyclooxygenase (COX) enzymes: COX-1 and COX-2. Our objective was to determine if long-term consumption of a flaxseed enriched diet decreased ovarian cancer severity and incidence in the laying hen and to investigate its potential correlation with the expression of COX enzymes and PGE2 concentration. METHODS: White Leghorn hens were fed 10% flaxseed-enriched or standard diet for 4years. The severity and incidence of ovarian cancer were determined by gross pathology and histology. COX-1 and COX-2 protein and mRNA expression and PGE2 concentrations in ovaries were measured by Western blot, quantitative real-time PCR and ELISA, respectively. RESULTS: The results demonstrated that there was a reduction in ovarian cancer severity and incidence in hens fed flaxseed diet. In correlation with decreased ovarian cancer severity and incidence, concentration of PGE2 and expression of COX-2 were diminished in ovaries of hens fed flaxseed. CONCLUSIONS: Our findings suggest that the lower levels of COX-2 and PGE2 are the main contributing factors in the chemo-suppressive role of long-term flaxseed consumption in ovarian cancer in laying hens. These findings may provide the basis for clinical trials of dietary intervention targeting prostaglandin biosynthesis for the prevention and treatment of ovarian cancer.


Assuntos
Dieta , Dinoprostona/biossíntese , Linho , Neoplasias Ovarianas/epidemiologia , Ovário/metabolismo , Sementes , Ração Animal , Animais , Galinhas , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Feminino , Incidência , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ovário/enzimologia , Ovário/patologia , RNA Mensageiro/metabolismo , Índice de Gravidade de Doença , Fatores de Tempo
3.
Lipids Health Dis ; 12: 152, 2013 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-24156238

RESUMO

BACKGROUND: An effective way to control cancer is by prevention. Ovarian cancer is the most lethal gynecological malignancy. Progress in the treatment and prevention of ovarian cancer has been hampered due to the lack of an appropriate animal model and absence of effective chemo-prevention strategies. The domestic hens spontaneously develop ovarian adenocarcinomas that share similar histological appearance and symptoms such as ascites and metastasis with humans. There is a link between chronic inflammation and cancer. Prostaglandin E2 (PGE2) is the most pro-inflammatory ecoisanoid and one of the downstream products of two isoforms of cyclooxygenase (COX) enzymes: COX-1 and COX-2. PGE2 exerts its effects on target cells by coupling to four subtypes of receptors which have been classified as EP1-4. Fish oil is a source of omega-3 fatty acids (OM-3FAs) which may be effective in prevention of ovarian cancer. Our objective was to assess the potential impact of fish oil on expression of COX enzymes, PGE2 concentration, apoptosis and proliferation in ovaries of laying hens. METHODS: 48 white Leghorn hens were fed 50, 100, 175, 375 and 700 mg/kg fish oil for 21 days. The OM3-FAs and omega-6 fatty acids contents of egg yolks were determined by Gas Chromatography. Proliferation, apoptosis, COX-1, COX-2 and prostaglandin receptor subtype 4 (EP4) protein and mRNA expression and PGE2 concentration in ovaries were measured by PCNA, TUNEL, Western blot, quantitative real-time qPCR and ELISA, respectively. RESULTS: Consumption of fish oil increased the incorporation of OM-3FAs into yolks and decreased both COX-1 and COX-2 protein and mRNA expression. In correlation with COXs down-regulation, fish oil significantly reduced the concentrations of PGE2 in ovaries. EP4 protein and mRNA expression in ovaries of hens was not affected by fish oil treatment. A lower dose of fish oil increased the egg laying frequency. 175 and 700 mg/kg fish oil reduced proliferation and 700 mg/kg increased apoptosis in hen ovaries. CONCLUSIONS: Our findings suggest that the lower doses of fish oil reduce inflammatory PG and may be an effective approach in preventing ovarian carcinogenesis. These findings may provide the basis for clinical trials utilizing fish oil as a dietary intervention targeting prostaglandin biosynthesis for the prevention and treatment of ovarian cancer.


Assuntos
Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Suplementos Nutricionais , Dinoprostona/antagonistas & inibidores , Óleos de Peixe/administração & dosagem , Neoplasias Epiteliais e Glandulares/prevenção & controle , Neoplasias Ovarianas/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Carcinoma Epitelial do Ovário , Proliferação de Células/efeitos dos fármacos , Galinhas , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/genética , Dinoprostona/biossíntese , Dinoprostona/genética , Gema de Ovo/química , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Ovário/efeitos dos fármacos , Ovário/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Zigoto/fisiologia
4.
Comp Med ; 71(4): 271-284, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34325771

RESUMO

Often referred to as the silent killer, ovarian cancer is the most lethal gynecologic malignancy. This disease rarely shows any physical symptoms until late stages and no known biomarkers are available for early detection. Because ovarian cancer is rarely detected early, the physiology behind the initiation, progression, treatment, and prevention of this disease remains largely unclear. Over the past 2 decades, the laying hen has emerged as a model that naturally develops epithelial ovarian cancer that is both pathologically and histologically similar to that of the human form of the disease. Different molecular signatures found in human ovarian cancer have also been identified in chicken ovarian cancer including increased CA125 and elevated E-cadherin expression, among others. Chemoprevention studies conducted in this model have shown that decreased ovulation and inflammation are associated with decreased incidence of ovarian cancer development. The purpose of this article is to review the major studies performed in laying hen model of ovarian cancer and discuss how these studies shape our current understanding of the pathophysiology, prevention, and treatment of epithelial ovarian cancer.


Assuntos
Galinhas , Neoplasias Ovarianas , Animais , Carcinoma Epitelial do Ovário , Feminino , Humanos , Neoplasias Ovarianas/prevenção & controle , Neoplasias Ovarianas/veterinária
5.
Gynecol Oncol ; 117(2): 341-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20153884

RESUMO

OBJECTIVE: With the exception of the laying hen, no other animal model of spontaneous ovarian surface epithelial cancer replicates the human disease. Flaxseed is the richest vegetable source of omega-3 fatty acids, which are chemopreventive in breast cancer and may be important in other cancers. The objective of this study was to determine if a flaxseed-enriched diet had a chemopreventive effect on ovarian cancer in the laying hen. METHODS: White Leghorn hens were fed with 10% flaxseed-enriched or standard diet for 1 year. The incidence and severity of ovarian cancer were determined by gross pathology and histology in the two groups. General health markers were also measured. Eggs were collected and analyzed by gas chromatography to determine omega-3 fatty acid levels. RESULTS: A significant reduction in late stage ovarian tumors was detected in the flaxseed-fed hens. Incidence rates of ovarian cancer were not significantly different between the two groups. The results indicate that a flaxseed diet increases overall survival in the laying hen. Flaxseed-fed hens' eggs incorporated significantly more omega-3 fatty acids compared to control hens. CONCLUSIONS: These findings show that 10% flaxseed supplementation for 1 year in the laying hen results in a significant reduction in the severity of ovarian cancer, but no change in the incidence of the disease. Hens fed flaxseed had overall better health and reduced mortality. These findings may provide the basis for a clinical trial that evaluates the efficacy of flaxseed as a chemosuppressant of ovarian cancer in women.


Assuntos
Ácidos Graxos Ômega-3/administração & dosagem , Linho , Neoplasias Ovarianas/dietoterapia , Animais , Galinhas , Dieta , Modelos Animais de Doenças , Feminino , Estadiamento de Neoplasias , Neoplasias Ovarianas/patologia , Distribuição Aleatória
6.
Oncotarget ; 11(40): 3646-3659, 2020 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-33088425

RESUMO

BACKGROUND: 2-methoxyestradiol (2MeOE2) is a natural metabolite of estradiol, which is generated by the action of CYP1A1 enzyme in the liver. We have previously shown that a flaxseed-supplemented diet decreases both the incidence and severity of ovarian cancer in laying hens, also induces CYP1A1 expression in liver. Recently, we have shown that as a biologically derived active component of flax diet, 2MeOE2 induces apoptosis in ovarian cancer cells which is partially dependent on p38 MAPK. The objective of this study was to elucidate the molecular mechanism of actions of 2MeOE2, a known microtubule disrupting agent, in inducing apoptosis in ovarian tumors. RESULTS: 2MeOE2 induces γH2Ax expression and apoptotic histone modifications in ovarian cancer cells, which are predicted downstream targets of protein kinase Cδ (PKCδ) during apoptosis. Overexpressing full length PKCδ alone does not induce apoptosis but potentiates 2MeOE2-mediated apoptosis. C3-domain mutated dominant-negative PKCδ (PKCδDN) significantly reduces 2MeOE2-induced caspase-3 cleavage and apoptotic histone modification. Silencing PKCδ diminishes 2MeOE2-mediated apoptosis. The catalytic fragment of PKCδ (PKCδCAT) evokes pro-apoptotic effects which are principally dependent on p38 MAPK phosphorylation. CONCLUSIONS: The pro-apoptotic actions of 2MeOE2 are in part dependent on catalytic activation of PKCδ. Catalytic activation of PKCδ accelerates the 2MeOE2-induced apoptotic cascade. This study describes a novel molecular action of flaxseed diet in ovarian cancer.

7.
Gynecol Oncol ; 113(3): 362-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19321195

RESUMO

OBJECTIVE: Epithelial ovarian carcinoma (EOC) is a leading cause of cancer deaths in women. Until recently, a significant lack of an appropriate animal model has hindered the discovery of early detection markers for ovarian cancer. The aging hen serves as an animal model because it spontaneously develops ovarian adenocarcinomas similar in histological appearance to the human disease. E-cadherin is an adherens protein that is down-regulated in many cancers, but has been shown to be up-regulated in primary human ovarian cancer. Our objective was to evaluate E-cadherin expression in the hen ovary and compare its expression to human ovarian cancer. METHODS: White Leghorn hens aged 185 weeks (cancerous and normal) were used for sample collection. A human ovarian tumor tissue array was used for comparison to the human disease. E-cadherin mRNA and protein expression were analyzed in cancerous and normal hen ovaries by immunohistochemistry (IHC), Western blot, and quantitative real-time PCR (qRT-PCR). Tissue fixed in neutral buffered formalin was used for IHC. Protein from tissue frozen in liquid nitrogen was analyzed by Western blot. RNA was extracted from tissue preserved in RNAlater and analyzed by qRT-PCR. The human ovarian tumor tissue array was used for IHC. RESULTS: E-cadherin mRNA and protein expression were significantly increased in cancerous hen ovaries as compared to ovaries of normal hens by qRT-PCR and Western blot. Similar expression of E-cadherin was observed by IHC in both human and hen ovarian cancer tissues. Similar E-cadherin expression was also observed in primary ovarian tumor and peritoneal metastatic tissue from cancerous hens. CONCLUSIONS: Our findings suggest that the up-regulation of E-cadherin is an early defining event in ovarian cancer and may play a significant role in the initial development of the primary ovarian tumor. E-cadherin also appears to be important in the development of secondary tumors within the peritoneal cavity. Our data suggest that E-cadherin may prove to be an important target in the preventative treatment of metastatic ovarian cancer and further confirm that the laying hen is a good model for the study of human epithelial ovarian carcinoma.


Assuntos
Caderinas/metabolismo , Neoplasias Ovarianas/metabolismo , Regulação para Cima , Animais , Galinhas , Modelos Animais de Doenças , Feminino , Humanos , Ovário/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
J Ovarian Res ; 12(1): 49, 2019 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-31128594

RESUMO

BACKGROUND: We have previously shown that a whole flaxseed supplemented diet decreased the onset and severity of ovarian cancer in the laying hen, the only known animal model of spontaneous ovarian cancer. Flaxseed is rich in omega-3 fatty acids (OM3FA), mostly α-Linoleic acid (ALA), which gets converted to Docosahexaenoic acid (DHA) by the action of delta-6 desaturase enzyme. Ingestion of flaxseed also causes an increase in production of 2-methoxyestradiol (2MeOE2) via the induction of the CYP1A1 pathway of estrogen metabolism. We have previously reported that the flaxseed diet induces apoptosis via p38-MAPK pathway in chicken tumors. The objective of this study was to investigate the effect of the flaxseed diet on ovarian cancer in chickens, focusing on two hallmarks of cancer, apoptosis and angiogenesis. RESULTS: The anti-cancer effects of two active biologically derived compounds of flax diet, 2MeOE2 and DHA, were individually tested on human ovarian cancer cells and in vivo by the Chick Chorioallantoic Membrane (CAM) assay. Our results indicate that a flaxseed-supplemented diet promotes apoptosis and inhibits angiogenesis in chicken tumors but not in normal ovaries. 2MeOE2 promotes apoptosis in human ovarian cancer cells, inhibits angiogenesis on CAM and its actions are dependent on the p38-MAPK pathway. DHA does not have any pro-apoptotic effect on human ovarian cancer cells but has strong anti-angiogenic effects as seen on CAM, but not dependent on the p38-MAPK pathway. CONCLUSIONS: Dietary flaxseed supplementation promotes a pro-apoptotic and anti-angiogenic effect in ovarian tumors, not in normal ovaries. The biologically derived active compounds from flaxseed diet act through different pathways to elicit their respective anti-cancer effects. A flaxseed-supplemented diet is a promising approach for prevention of ovarian cancer as well as having a significant potential as an adjuvant treatment to supplement chemotherapeutic agents for treatment of advanced stages of ovarian cancer.


Assuntos
2-Metoxiestradiol/farmacologia , Apoptose/efeitos dos fármacos , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/farmacologia , Linho , Neoplasias Ovarianas/prevenção & controle , 2-Metoxiestradiol/administração & dosagem , Animais , Linhagem Celular Tumoral , Galinhas , Membrana Corioalantoide , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/administração & dosagem , Feminino , Linho/química , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neovascularização Patológica/prevenção & controle , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ovário , Sementes/química
9.
Gynecol Oncol ; 109(1): 115-21, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18272210

RESUMO

OBJECTIVE: Reduced Selenium-Binding Protein 1 (SELENBP1) expression was recently shown in multiple cancers. There is little information on the expression and function of SELENBP1 in cancer progression. In order to develop a better understanding of the role of SELENBP1 in ovarian cancer, our objective was to determine if SELENBP1 is expressed in the normal ovaries and ovarian tumors in the egg-laying hen, a spontaneous model of human ovarian cancer. METHODS: SPB1 mRNA expression in normal ovary (n=20) and ovarian tumors (n=23) was evaluated by RT-PCR. Relative levels of mRNA were compared by quantitative RT-PCR (qRT-PCR) in selected samples. SELENBP1 protein expression was evaluated by 1D Western blot and immunohistochemistry with a commercial anti-human SELENBP1 antibody. RESULTS: SELENBP1 mRNA and protein was expressed in 100% of normal and ovarian tumors and qRT-PCR confirmed decreased mRNA expression in 80% of ovarian tumors. SELENBP1 was primarily localized in surface epithelial cells of normal ovaries. In ovaries containing early tumor lesions, SELENBP1 expression was reduced in the surface epithelium near the tumor and was expressed in tumor cells, while more distant regions with normal histology retained SELENBP1 expression in the surface epithelium. CONCLUSIONS: We have shown for the first time that SELENBP1 is expressed in both normal ovaries and ovarian tumors in the hen and that SELENBP1 expression is altered in the vicinity of the tumor. Furthermore, SELENBP1 expression in normal ovarian surface epithelium and in ovarian tumors parallels that previously reported for ovarian cancer in women.


Assuntos
Adenocarcinoma/metabolismo , Galinhas , Modelos Animais de Doenças , Neoplasias Ovarianas/metabolismo , Proteínas de Ligação a Selênio/biossíntese , Adenocarcinoma/genética , Animais , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Ovarianas/genética , Ovário/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas de Ligação a Selênio/genética
10.
J Nutr Biochem ; 42: 117-125, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28178600

RESUMO

The study reported here demonstrates that a flaxseed-supplemented diet causes ovarian tumors in the laying hen to undergo apoptosis, resulting in a reduction of tumor burden, reducing the frequency and severity of ovarian cancer. We have previously shown in normal ovaries that flaxseed and its components down-regulate ERalpha and alter the expression of enzymes that metabolize estrogen. In this study, we analyzed the effects of the two main components of whole flaxseed, ligan and omega 3 fatty acids on estrogen metabolism and the estrogen receptor in ovarian tumors. ER alpha expression was up-regulated in the ovarian tumors and was not affected by diet. Liver CYP1A1 expression was significantly increased by the whole flaxseed diet with a corresponding increase in 2-methoxyestradiol plasma levels. We also observed increased p38 and ERK 1/2 MAPK activation in the ovary as well as an increase in apoptosis in the tumor epithelium. SMAD 7, a factor involved in the 2-methoxyestradiol-mediated apoptosis pathway was also up-regulated in tumors from the whole flaxseed diet group. 2-methoxyestradiol-induced antitumor effects were further validated by in human ovarian cancer cells. This study details the effect of flaxseed diet on estrogen metabolism and demonstrates the antiovarian cancer effects of 2-methoxyestradiol.


Assuntos
Linho , Neoplasias Ovarianas/dietoterapia , Ovário/metabolismo , 2-Metoxiestradiol , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular Tumoral , Galinhas , Citocromo P-450 CYP1B1/metabolismo , Citocromo P-450 CYP3A/metabolismo , Suplementos Nutricionais , Estradiol/análogos & derivados , Estradiol/metabolismo , Estradiol/toxicidade , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Neoplasias Ovarianas/induzido quimicamente , Neoplasias Ovarianas/patologia , Proteína Smad7/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
J Steroid Biochem Mol Biol ; 159: 73-85, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26925929

RESUMO

Flaxseed has been studied for decades for its health benefits that include anti-cancer, cardio-protective, anti-diabetic, anti-inflammatory properties. The biologically active components that mediate these effects are the omega-3 fatty acids and the lignan, secoisolariciresinol diglucoside. We have previously shown that whole flaxseed supplemented diet decreases the severity and incidence of ovarian cancer while a 15% dose of flaxseed is most protective against inflammation and estrogen-induced chemical and genotoxicity. The objective of this study was to dissect the independent effects of the two flaxseed components on estrogen signaling and metabolism. Two and half year old hens were fed either a control diet, 15% whole flaxseed diet, defatted flax meal diet or 5% flax oil diet for 3 months after which the animals were sacrificed and blood and tissues were harvested. Whole flaxseed diet caused a decrease in expression of ERα. ERα target gene expression was assessed using RT(2) profiler PCR array. Some targets involved in the IGF/insulin signaling pathway (IRS1, IGFBP4, IGFBP5) were downregulated by flaxseed and its components. Flaxseed diet also downregulated AKT expression. A number of targets related to NF-kB signaling were altered by flaxseed diet including a series of targets implicated in cancer. Whole flaxseed diet also affected E2 metabolism by increasing CYP1A1 expression with a corresponding increase in the onco-protective E2 metabolite, 2-methoxyestradiol. The weak anti-estrogens, enterolactone, enterodiol and 2-methoxyestradiol, might be working synergistically to generate a protective effect on the ovaries from hens on whole flaxseed diet by altering the estrogen signaling and metabolism.


Assuntos
Suplementos Nutricionais , Óleo de Semente do Linho/administração & dosagem , Neoplasias Ovarianas/veterinária , Lesões Pré-Cancerosas/veterinária , 2-Metoxiestradiol , Animais , Proteínas Aviárias/metabolismo , Galinhas , Citocromo P-450 CYP1A1/metabolismo , Estradiol/análogos & derivados , Estradiol/sangue , Feminino , Linho/química , Expressão Gênica , Fígado/enzimologia , NF-kappa B/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/prevenção & controle , Ovário/enzimologia , Lesões Pré-Cancerosas/dietoterapia , Lesões Pré-Cancerosas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais
12.
Endocrinology ; 144(7): 2882-91, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12810543

RESUMO

Reactive oxygen species (ROS) are involved in a variety of pathophysiological conditions of the testis, and oxidative stress is known to inhibit ovarian and testicular steroidogenesis. The site of ROS-mediated inhibition of steroidogenesis in the corpus luteum and MA-10 tumor Leydig cells was shown to be the hormone-sensitive mitochondrial cholesterol transfer step. The purpose of this study was to examine the effects of ROS on steroidogenic acute regulatory (StAR) protein in MA-10 cells and determine the extent to which MA-10 cell mitochondria are sensitive to oxidative stress. cAMP-stimulated progesterone production was inhibited in a dose-dependent manner in MA-10 cells exposed to H(2)O(2). StAR protein, but not mRNA levels, was decreased in parallel to changes in progesterone production. Even at the highest concentrations of H(2)O(2) tested, there was no effect on P450 side-chain cleavage enzyme protein levels. Oxidative stress from exposure to exogenous xanthine oxidase and xanthine resulted in the inhibition of both progesterone production and StAR protein expression. The mature 30- and 32-kDa intramitochondrial forms of StAR were decreased relative to the 37-kDa extramitochondrial precursor form of StAR, indicating that the ROS-mediated inhibition of StAR protein was due, in part, to the inhibition of mitochondrial import and processing. Vital staining with the fluorescent dye tetramethylrhodamine ethyl ester was used to visualize changes in the mitochondrial electrochemical gradient-dependent membrane potential (Deltapsim). ROS caused a significant dissipation of Deltapsi(m) and time-dependent loss of tetramethylrhodamine ethyl ester fluorescence. The inhibitory effects of H(2)O(2) were transient. There was no evidence for ROS-induced cell death, and following H(2)O(2) removal in the presence of continuous treatment with 8-bromo-cAMP, StAR protein levels and progesterone production were restored. In addition, there was no loss of cell viability following treatment with H(2)O(2) or xanthine/xanthine oxidase as determined by trypan blue exclusion. H(2)O(2) did not cause a significant decrease in total cellular ATP levels. These data indicate that oxidative stress-mediated perturbation of the mitochondria and dissipation of Deltapsi(m) results in the inhibition of StAR protein expression and its import, processing, and cholesterol transfer activity. These findings confirm earlier studies demonstrating the requirement for maintenance of an intact Deltapsi(m) for StAR protein function in cholesterol transport. The significant reduction in the 32- to 30-kDa mature forms of StAR, cessation of cholesterol transport, and loss of Deltapsi(m) are consistent with mitochondrial perturbation because of oxidative stress. This mechanism likely contributes to a host of pathophysiological events evident in testicular disorders such as infection, reperfusion injury, aging, cryptorchidism, and varicocele.


Assuntos
Células Intersticiais do Testículo/metabolismo , Mitocôndrias/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Progesterona/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , AMP Cíclico/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Peróxido de Hidrogênio/farmacologia , Células Intersticiais do Testículo/citologia , Masculino , Potenciais da Membrana/fisiologia , Camundongos , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas , Xantina Oxidase/metabolismo
13.
Endocrinology ; 143(8): 2986-94, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12130564

RESUMO

LH receptor activation leads to the phosphorylation/activation of p42/44 MAPK in preovulatory granulosa cells. As the LH receptor can activate both adenylyl cyclase and phospholipase C, we hypothesized that the LH receptor could elicit phosphorylation of p42/44 MAPK through activation of protein kinase A (PKA) and/or protein kinase C (PKC). Preovulatory granulosa cells in serum-free primary cultures were treated with ovulatory concentrations of human chorionic gonadotropin (hCG), an LH receptor agonist, with or without various inhibitors. The PKA inhibitor H89 as well as the myristoylated PKA inhibitor peptide PKI strongly inhibited hCG-stimulated p42/44 MAPK phosphorylation, whereas the PKC inhibitor GF109203X had no effect on p42/44 MAPK phosphorylation. LH receptor-stimulated phosphorylation of cAMP response element-binding protein (CREB), histone H3, and MAPK kinase (MEK) was also strongly inhibited by H89 and not by GF109203X. The extent of PKC activation was assessed in preovulatory granulosa cells using three criteria: translocation of PKC isoforms to the membrane fraction, phosphorylation of a known PKC substrate, and autophosphorylation of PKC delta on an activation-related site. By all three criteria PKCs were partially activated before hCG stimulation, and hCG treatment failed to elicit further PKC activation, in vitro or in vivo. Taken together, these results indicate that, under primary culture conditions where physiological levels of signaling proteins are present, hCG signals to activate MEK, p42/44 MAPK, CREB, and histone H3 in a predominantly PKA-dependent and PKC-independent manner. Unexpectedly, PKCs were partially activated in the absence of LH receptor activation, and LH receptor activation did not elicit further detectable PKC activation.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , MAP Quinase Quinase Quinase 1 , Proteínas de Membrana , Proteína Quinase C/fisiologia , Receptores do LH/fisiologia , Animais , Células Cultivadas , Gonadotropina Coriônica/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Ativação Enzimática , Glucosidases , Histonas/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Substrato Quinase C Rico em Alanina Miristoilada , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley
14.
Mol Cell Endocrinol ; 215(1-2): 127-34, 2004 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-15026185

RESUMO

Post-translational modifications such as phosphorylation and specific proteolysis affect the steroidogenic acute regulatory protein (StAR) activity. We have found that in pcDNA3.1-StAR-transfected COS-1 cells, StAR was phosphorylated on S55, S56 and S194 (Fleury et al., unpublished). In this study, we are comparing the two-dimensional gel electrophoresis (2D-PAGE) characteristics of the WT StAR with those of the S194A and S55A/S56A/S194A-StAR mutants under control and (Bu)(2)-cAMP stimulation, using an anti-StAR antibody and an anti-phospho-(Ser/Thr) PKA substrate antibody. The 2D-PAGE migration pattern of the WT StAR analyzed by immunoblotting with the anti-StAR antibody revealed many StAR species with different pI and different molecular weights. In the (Bu)(2)-cAMP-WT preparations, except for three, all these StAR species were also recognized by the anti-phospho-(Ser/Thr) PKA substrate antibody; in contrast, less phosphorylated species were found in the non-stimulated WT preparations. The two-dimensional (2D) patterns of StAR revealed by the anti-StAR and the anti-phospho-(Ser/Thr) PKA substrate antibodies were modified for the S194A mutant and further modified for the S55A/S56A/S194A mutant. Whereas many species could still be detected by the anti-StAR antibody in the triple mutant S55A/S55A/S194A, none of these could be revealed by the anti-phospho-(Ser/Thr) PKA substrate antibody. Finally we found that, in addition to phosphorylation, the formation of different StAR species was also due to the hydrolysis of the molecule at its N-terminal and to a lesser degree at its C-terminal.


Assuntos
Córtex Suprarrenal/metabolismo , Mutação/genética , Fosfoproteínas/metabolismo , Processamento de Proteína Pós-Traducional , Córtex Suprarrenal/citologia , Córtex Suprarrenal/efeitos dos fármacos , Animais , Bucladesina/farmacologia , Células COS , Cricetinae , Eletroforese em Gel Bidimensional , Immunoblotting , Fosfoproteínas/genética , Fosforilação
15.
J Reprod Immunol ; 57(1-2): 3-18, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12385830

RESUMO

This review will highlight recent advances in the study of the immuno-endocrinology of the testis, in particular how macrophage-derived inflammatory mediators affect Leydig cell functions. Both the beneficial and deleterious outcomes resulting from macrophage-Leydig cell interactions are discussed. A brief overview of testicular physiology is provided that discusses the functional and anatomical compartmentalization of the testis into the gamete and endocrine compartments where spermatogenesis and testosterone biosynthesis take place, respectively. The process of steroidogenesis including the activities of the steroidogenic enzymes and the role of steroidogenic acute regulatory protein (StAR) are described. The close physical association between Leydig cells and interstitial testicular macrophages suggests that these cells are functionally related. Under normal physiological and non-inflammatory conditions macrophages play an important role in Leydig cell development. If macrophages are absent from the testicular interstitium, Leydig cells fail to develop normally, which suggest that macrophages provide essential growth and differentiation factors for Leydig cells. In contrast, when macrophages are activated and elaborate inflammatory mediators, Leydig cell steroidogenesis is inhibited. Activated macrophages produce pro-inflammatory cytokines such as interleukin-1 (IL-1) and tumor necrosis factor (TNF) that are profoundly inhibitory to Leydig cells and appear to act as transcriptional repressors of steroidogenic enzyme gene expression. Macrophages also produce reactive oxygen species (ROS) such as hydrogen peroxide, which also inhibits Leydig cell functions. ROS appear to act acutely by perturbing Leydig cell mitochondria resulting in the inhibition of StAR protein expression. One important consequence of this immune modulation of Leydig cell function may be manifest behaviorally by switching the affected animal from 'testosterone' behavior, to 'sickness' behavior. Increased interest in immune-endocrine control of reproductive function over the past decade has stimulated research into the molecular and biochemical immunopathophysiology of the reproductive system. As investigations unravel mechanisms underlying reproductive dysfunction caused by inflammation and infection, an understanding of the role that immune-endocrine interactions play in the normal physiology of the reproductive system has emerged.


Assuntos
Células Intersticiais do Testículo/metabolismo , Macrófagos/imunologia , Esteroides/biossíntese , Testículo/imunologia , Testículo/fisiologia , Animais , Comunicação Celular , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Espécies Reativas de Oxigênio/metabolismo , Testículo/citologia , Testosterona/biossíntese
16.
J Steroid Biochem Mol Biol ; 91(4-5): 259-71, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15336703

RESUMO

In order to study the effect of phosphorylation on the function of the steroidogenic acute regulatory protein (StAR), 10 putative phosphorylation sites were mutated in the hamster StAR. In pcDNA3.1-StAR transfected COS-1 cells, decreases in basal activity were found for the mutants S55A, S185A and S194A. Substitution of S185 by D or E to mimic phosphorylation resulted in decreased activity for all mutants; we concluded that S185 was not a phosphorylation site and we hypothesized that mutations on S185 created StAR conformational changes resulting in a decrease in its binding affinity for cholesterol. In contrast, the mutation S194D resulted in an increase in StAR activity. We have calculated the relative rate of pregnenolone formation (App. V(max)) in transfected COS-1 cells with wild type (WT) and mutant StAR-pcDNA3.1 under control and (Bu)(2)-cAMP stimulation. The App. V(max) values refer to the rate of cholesterol transported and metabolized by the cytochrome P450scc enzyme present in the inner mitochondrial membrane. The App. V(max) was 1.61 +/- 0.28 for control (Ctr) WT StAR and this value was significantly increased to 4.72 +/- 0.09 for (Bu)(2)-cAMP stimulated preparations. App. V(max) of 5.53 (Ctr) and 4.82 ((Bu)(2)-cAMP) found for S194D StAR preparations were similar to that of the WT StAR stimulated preparations. At equal StAR quantity, an anti-phospho-(S/T) PKA substrate antibody revealed four times more phospho-(S/T) in (Bu)(2)-cAMP than in control preparations. The intensity of phosphorylated bands was decreased for the S55A, S56A and S194A mutants and it was completely abolished for the S55A/S56A/S194A mutant. StAR activity of control and stimulated preparations were diminished by 73 and 72% for the mutant S194A compared to 77 and 83% for the mutant S55A/S56A/S194A. The remaining activity appears to be independent of phosphorylation at PKA sites and could be due to the intrinsic activity of non-phosphorylated StAR or to an artefact due to the pharmacological quantity of StAR expressed in COS-1. In conclusion we have shown that (Bu)(2)-cAMP provokes an augmentation of both the quantity and activity of StAR, and that an enhancement in StAR phosphorylation increases its activity. The increased quantity of StAR upon (Bu)(2)-cAMP stimulation could be due to an augmentation of its mRNA or protein synthesis stability, or both; this is yet to be determined.


Assuntos
Glândulas Suprarrenais/metabolismo , Mutação/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Animais , Sítios de Ligação , Bucladesina/farmacologia , Células COS , Chlorocebus aethiops , Colesterol/metabolismo , Enzima de Clivagem da Cadeia Lateral do Colesterol , Cricetinae , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mitocôndrias , Mutagênese Sítio-Dirigida , Fosforilação , Pregnenolona/metabolismo , Ligação Proteica , Conformação Proteica/efeitos dos fármacos , Transfecção
17.
Artigo em Inglês | MEDLINE | ID: mdl-23978451

RESUMO

Prevention of ovarian cancer is the best approach for reducing the impact of this deadly disease. The laying hen is a robust model of spontaneous ovarian cancer that recapitulates the human disease. Dietary intervention with flaxseed, the richest vegetable source of omega-3 fatty acids (OM-3FAs) and phytoestrogen lignans, demonstrate the potential for effective prevention and amelioration of ovarian cancer by targeting inflammatory prostaglandin pathways. Prostaglandin E2 (PGE2) is the most pro-inflammatory ecoisanoid and one of the downstream products of two isoforms of cyclooxygenase (COX) enzymes: COX-1 and COX-2. Our objective was to investigate the effect of flaxseed supplementation for one year on ovarian cancer and correlate its effects to expression of COX enzymes and concentrations of prostaglandins. White Leghorn hens were fed 10% flaxseed-enriched or standard diet for one year. The severity of ovarian cancer was determined by gross pathology and histology. COX-1 and COX-2 localization and protein and mRNA expression and PGE2 and PGE3 concentrations in ovaries were measured by IHC, western blot, quantitative real-time PCR and LC-MS-MS, respectively. The results demonstrated a significant reduction in late stage ovarian tumors in the flaxseed-fed hens compared with the control diet-fed hens. In correlation with decreased ovarian cancer severity, concentrations of PGE2 and expression of COX-2 were diminished in ovaries of flaxseed-fed hens. PGE3 concentrations were below the level of detection. The results demonstrated that in normal ovaries, COX-1 was localized to the granulosa cell layer surrounding the follicles and ovarian surface epithelium (OSE) whereas COX-2 protein was localized to the granulosa cell layer in the follicle. Extensive COX-1 and COX-2 protein expression was found throughout the ovarian carcinoma. Our findings suggest that the flaxseed-mediated reduction in the severity of ovarian cancer in hens is correlated to the reduction in PGE2 in the ovaries of flaxseed-fed hens. These findings may provide the basis for clinical trials of dietary intervention targeting prostaglandin biosynthesis for the prevention and treatment of ovarian cancer.


Assuntos
Dinoprostona/biossíntese , Linho , Neoplasias Ovarianas/veterinária , Ovário/metabolismo , Doenças das Aves Domésticas/prevenção & controle , Ração Animal , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Galinhas , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dieta , Suplementos Nutricionais , Ácidos Graxos Ômega-3/metabolismo , Feminino , Expressão Gênica , Humanos , Neoplasias Ovarianas/prevenção & controle , Ovulação , Receptores de Prostaglandina E Subtipo EP4/genética
18.
Artigo em Inglês | MEDLINE | ID: mdl-23089186

RESUMO

Chronic inflammation has been linked to cancer. Prostaglandin E2 (PGE2) is the most pro-inflammatory lipid and one of the downstream products of 2 isoforms of cyclooxygenase (COX) enzymes: COX-1 and COX-2. Ovarian cancer is the most lethal gynecological malignancy and mainly occurs in older women. The factors that contribute to the correlation of age and ovarian cancer are unknown. The purpose of this study was to examine the expression of COX enzymes and PGE2 levels in ovaries and correlate them to ovarian cancer and aging. White Leghorn hens aged 1, 1.5, 2, 2.5, 3 and 3.5 years were used. The incidence of ovarian cancer was determined by gross pathology and histology. COX-1 and COX-2 protein and mRNA expression and PGE2 concentrations in ovaries were measured using Western blot, quantitative real-time PCR and ELISA, respectively. Our results indicated an increase in ovarian cancer incidence and expression of both COX enzymes in ovaries of older hens. In correlation with ovarian cancer incidence and COX enzymes expression, PGE2 concentrations were elevated with age. Ovaries with tumor had elevated COX-1 expression and PGE2 concentration compared to normal ovaries. Our findings suggest that the up-regulation of COX enzymes with age is the main contributing factor in the age associated increase in PGE2. Furthermore, elevated PGE2 in ovaries of hens concomitant with age suggests its important role in early stages of ovarian carcinogenesis. These finding may provide the basis for clinical trials utilizing COX specific inhibitors or dietary intervention targeting prostaglandin biosynthesis for the prevention and treatment of ovarian cancer.


Assuntos
Adenocarcinoma/veterinária , Envelhecimento , Neoplasias Ovarianas/veterinária , Ovário/metabolismo , Doenças das Aves Domésticas/metabolismo , Prostaglandinas/metabolismo , Regulação para Cima , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Animais Endogâmicos , Proteínas Aviárias/biossíntese , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Galinhas , Ciclo-Oxigenase 1/biossíntese , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/biossíntese , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Indução Enzimática , Feminino , Regulação Neoplásica da Expressão Gênica , Illinois , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ovário/patologia , RNA Mensageiro/metabolismo
19.
Endocrine ; 33(3): 235-44, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18498063

RESUMO

Cyclooxygenase (COX) (PTGS) is the rate-limiting enzyme in the biosynthesis of prostaglandins. Two COX isoforms have been identified, COX-1 and COX-2, which show distinct cell-specific expression and regulation. Ovarian cancer is the most lethal gynecological malignancy and the disease is poorly understood due to the lack of suitable animal models. The laying hen spontaneously develops epithelial ovarian cancer with few or no symptoms until the cancer has progresses to a late stage, similar to the human disease. The purpose of this study was to examine the relative expression and distribution of COX-1 and COX-2 in the ovaries of normal hens and in hens with ovarian cancer. The results demonstrate that COX-1 was localized to the granulosa cell layer and cortical interstitium, ovarian surface epithelium (OSE) and postovulatory follicle (POF) of the normal ovary. In ovarian cancer, COX-1 mRNA was significantly increased and COX-1 protein was broadly distributed throughout the tumor stroma. COX-2 protein was localized to the granulosa cell layer in the follicle and the ovarian stroma. COX-2 mRNA expression did not change as a function of age or in ovarian cancer. There was significantly higher expression of COX-1 mRNA in the first POF (POF-1) compared to POF-2 and POF-3. COX-2 mRNA expression was not significantly different among POFs. There was no difference in COX-1 or COX-2 mRNA in the OSE isolated from individual follicles in the follicular hierarchy. The results confirm previous findings of the high expression of COX-1 in ovarian tumors further supporting the laying hen as a model for ovarian cancer, and demonstrate for the first time the high expression of COX-1 in POF-1 which is the source of prostaglandins needed for oviposition.


Assuntos
Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/genética , Folículo Ovariano/enzimologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Animais , Galinhas , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Folículo Ovariano/patologia , Neoplasias Ovarianas/patologia , Oviposição , Ovulação , RNA Mensageiro/metabolismo , Regulação para Cima/fisiologia
20.
Glia ; 45(3): 213-28, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14730695

RESUMO

Steroidogenic acute regulatory protein (StAR) plays a key role in the availability of cholesterol to the inner mitochondrial membrane, where the first step of steroidogenesis, its conversion to pregnenolone, takes place. Here, we demonstrate for the first time that the StAR gene is also expressed in the rat sciatic nerve and in cultured Schwann cells. The addition to the culture medium of the cAMP-elevating agent forskolin or of the cAMP analogue 8Br-cAMP produced a time-course extinction of StAR gene expression. An inverse relationship was demonstrated between StAR gene expression and the intracellular cAMP content. Accordingly, pharmacological inhibition of the activities of Schwann cell adenylyl cyclase or of phosphodiesterase IV resulted in modifications of StAR gene expression. Since StAR gene expression is stimulated by cAMP in classical steroidogenic cells, our work is the first demonstration of a negative regulation of StAR gene by cAMP.


Assuntos
AMP Cíclico/fisiologia , Regulação para Baixo/fisiologia , Regulação da Expressão Gênica/fisiologia , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Células de Schwann/metabolismo , Animais , Células Cultivadas , Colforsina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Fosfoproteínas/antagonistas & inibidores , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Células de Schwann/efeitos dos fármacos , Nervo Isquiático/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA