Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Trace Elem Med Biol ; 79: 127216, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37224746

RESUMO

BACKGROUND: Apoptotic and oxido-inflammatory pathways have been found to be up-regulated in lead acetate poisoning which has been associated to endothelial and testicular dysfunctions. It is yet uncertain, nevertheless, if treatment with Ginkgo biloba supplements (GBS), a flavonoid-rich natural product can lessen the adverse effects of lead on endothelial and testicular functions. This study investigated the impact of Ginkgo biloba supplementation on lead-induced endothelial and testicular dysfunctions. METHODS: The animals were treated with GBS (50 mg/kg and 100 mg/kg orally) for 14 days following oral exposure to lead acetate (25 mg/kg) for 14 days. After euthanasia, blood samples, epididymal sperm, testes, and aorta were collected. The quantities of the hormones (testosterone, follicle stimulating hormone (FSH) and luteinizing hormone (LH), as well as the anti-apoptotic, oxidative, nitrergic, inflammatory markers, were then determined using immunohistochemistry, ELISA, and conventional biochemical methods. RESULTS: GBS reduced lead-induced oxidative stress by increasing the levels of the antioxidant enzymes catalase (CAT), glutathione (GSH), and superoxide dismutase (SOD), while lowering malondialdehyde (MDA) in endothelium and testicular cells. Normal testicular weight was restored by GBS which also decreased endothelial endothelin-I and increased nitrite levels. TNF-α and IL-6 were decreased while Bcl-2 protein expression was enhanced. Lead-induced alterations in reproductive hormones (FSH, LH, and testosterone) were also restored to normal. CONCLUSION: According to our result, using Ginkgo biloba supplement prevented lead from causing endothelial and testicular dysfunction by raising pituitary-testicular hormone levels, boosting Bcl-2 protein expression and lowering oxidative and inflammatory stress in the endothelium and testes.


Assuntos
Hormônios Testiculares , Testículo , Ratos , Animais , Masculino , Ratos Wistar , Ginkgo biloba/metabolismo , Regulação para Baixo , Regulação para Cima , Hormônios Testiculares/metabolismo , Hormônios Testiculares/farmacologia , Chumbo/metabolismo , Antioxidantes/metabolismo , Testosterona , Estresse Oxidativo , Hormônio Foliculoestimulante/metabolismo , Hormônio Foliculoestimulante/farmacologia , Glutationa/metabolismo , Suplementos Nutricionais , Sementes/metabolismo
2.
Horm Behav ; 73: 135-41, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26159287

RESUMO

Previous research has shown that exposure to testicular hormones during the peri-pubertal period of life has long-term, organizational effects on adult sexual behaviour and underlying neural mechanisms in laboratory rodents. However, the organizational effects of peri-pubertal testicular hormones on other aspects of behaviour and brain function are less well understood. Here, we investigated the effects of manipulating peri-pubertal testicular hormone exposure on later behavioural responses to novel environments and on hormone receptors in various brain regions that are involved in response to novelty. Male rodents generally spend less time in the exposed areas of novel environments than females, and this sex difference emerges during the peri-pubertal period. Male Lister-hooded rats (Rattus norvegicus) were castrated either before puberty or after puberty, then tested in three novel environments (elevated plus-maze, light-dark box, open field) and in an object/social novelty task in adulthood. Androgen receptor (AR), oestrogen receptor (ER1) and corticotropin-releasing factor receptor (CRF-R2) mRNA expression were quantified in the hypothalamus, hippocampus and medial amygdala. The results showed that pre-pubertally castrated males spent more time in the exposed areas of the elevated-plus maze and light-dark box than post-pubertally castrated males, and also confirmed that peri-pubertal hormone exposure influences later response to an opposite-sex conspecific. Hormone receptor gene expression levels did not differ between pre-pubertally and post-pubertally castrated males in any of the brain regions examined. This study therefore demonstrates that testicular hormone exposure during the peri-pubertal period masculinizes later response to novel environments, although the neural mechanisms remain to be fully elucidated.


Assuntos
Comportamento Exploratório/efeitos dos fármacos , Maturidade Sexual/efeitos dos fármacos , Comportamento Social , Hormônios Testiculares/farmacologia , Animais , Hormônio Liberador da Corticotropina/metabolismo , Feminino , Hipocampo/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Ratos , Receptores Androgênicos/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de Estrogênio/metabolismo , Maturidade Sexual/fisiologia
3.
Reprod Biol Endocrinol ; 9(1): 128, 2011 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-21942998

RESUMO

BACKGROUND: Spermatozoa leaving the testis are not able to fertilize the egg in vivo. They must undergo further maturation in the epididymis. Proteins secreted to the epididymal lumen by the epithelial cells interact with the spermatozoa and enable these maturational changes, and are responsible for proper storage conditions before ejaculation. The present study was carried out in order to characterize the expression of a novel Pate (prostate and testis expression) gene family, coding for secreted cysteine-rich proteins, in the epididymis. METHODS: Murine genome databases were searched and sequence comparisons were performed to identify members of the Pate gene family, and their expression profiles in several mouse tissues were characterized by RT-PCR. Alternate transcripts were identified by RT-PCR, sequencing and Northern hybridization. Also, to study the regulation of expression of Pate family genes by the testis, quantitative (q) RT-PCR analyses were performed to compare gene expression levels in the epididymides of intact mice, gonadectomized mice, and gonadectomized mice under testosterone replacement treatment. RESULTS: A revised family tree of Pate genes is presented, including a previously uncharacterized Pate gene named Pate-X, and the data revealed that Acrv1 and Sslp1 should also be considered as members of the Pate family. Alternate splicing was observed for Pate-X, Pate-C and Pate-M. All the Pate genes studied are predominantly expressed in the epididymis, whereas expression in the testis and prostate is notably lower. Loss of androgens and/or testicular luminal factors was observed to affect the epididymal expression of several Pate genes. CONCLUSIONS: We have characterized a gene cluster consisting of at least 14 expressed Pate gene members, including Acrv1, Sslp1 and a previously uncharacterized gene which we named Pate-X. The genes code for putatively secreted, cysteine-rich proteins with a TFP/Ly-6/uPAR domain. Members of the Pate gene cluster characterized are predominantly expressed in the murine epididymis, not in the testis or prostate, and are regulated by testicular factors. Similar proteins are present in venoms of several reptiles, and they are thought to mediate their effects by regulating certain ion channels, and are thus expected to have a clinical relevance in sperm maturation and epididymal infections.


Assuntos
Androgênios/farmacologia , Epididimo/metabolismo , Proteínas de Membrana/genética , Hormônios Testiculares/farmacologia , Testículo/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Epididimo/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Dados de Sequência Molecular , Família Multigênica/genética , Família Multigênica/fisiologia , Especificidade de Órgãos/genética , Filogenia , Proteínas de Plasma Seminal/genética , Proteínas de Plasma Seminal/metabolismo , Homologia de Sequência , Testículo/fisiologia
4.
Cancer Res ; 67(6): 2747-56, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363596

RESUMO

Mullerian-inhibiting substance (MIS), a transforming growth factor-beta family member, activates the nuclear factor-kappaB (NF-kappaB) pathway and induces the expression of B-cell translocation gene 2 (BTG2), IFN regulatory factor-1 (IRF-1), and the chemokine Gro-beta. Inhibiting NF-kappaB activation with a phosphorylation-deficient IkappaBalpha mutant abrogated MIS-mediated induction of all three genes. Expression of dominant-negative Smad1, in which serines at the COOH-terminal SSVS motif are converted to alanines, suppressed MIS-induced Smad1 phosphorylation and impaired MIS-stimulated Gro-beta promoter-driven reporter expression and Gro-beta mRNA. Suppressing Smad1 expression using small interfering RNA also mitigated MIS-induced Gro-beta mRNA, suggesting that regulation of Gro-beta expression by MIS was dependent on activation of NF-kappaB as well as Smad1. However, induction of IRF-1 and BTG2 mRNAs by MIS was independent of Smad1 activation. Characterization of kappaB-binding sequences within Gro-beta, BTG2, and IRF-1 promoters showed that MIS stimulated binding of p50 and p65 subunits to all three sites, whereas phosphorylated Smad1 (phospho-Smad1) protein was detectable only in the NF-kappaB complex bound to the kappaB site of the Gro-beta promoter. Consistent with these observations, chromatin immunoprecipitation assays showed recruitment of both phospho-Smad1 and p65 to the Gro-beta promoter in vivo, whereas p65, but not phospho-Smad1, was recruited to the BTG2 promoter. These results show a novel interaction between MIS-stimulated Smad1 and NF-kappaB signaling in which enhancement of NF-kappaB DNA binding and gene expression by phospho-Smad1 is dependent on the sequence of the kappaB consensus site within the promoter.


Assuntos
Neoplasias da Mama/metabolismo , Quimiocinas CXC/biossíntese , Glicoproteínas/farmacologia , NF-kappa B/metabolismo , Proteína Smad1/metabolismo , Hormônios Testiculares/farmacologia , Hormônio Antimülleriano , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Quimiocina CXCL2 , Quimiocinas CXC/genética , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Proteínas Imediatamente Precoces/biossíntese , Proteínas Imediatamente Precoces/genética , Fator Regulador 1 de Interferon/biossíntese , Fator Regulador 1 de Interferon/genética , NF-kappa B/antagonistas & inibidores , Fosforilação , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/antagonistas & inibidores , Proteína Smad1/biossíntese , Proteína Smad1/genética , Fator de Transcrição RelA/metabolismo , Proteínas Supressoras de Tumor
6.
Proc Natl Acad Sci U S A ; 103(30): 11154-9, 2006 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-16849428

RESUMO

The recent identification of "side population" (SP) cells in a number of unrelated human cancers and their normal tissue sources has renewed interest in the hypothesis that cancers may arise from somatic stem/progenitor cells. The high incidence of recurrence attributable to multidrug resistance and the multiple histologic phenotypes indicative of multipotency suggests a stem cell-like etiology of ovarian cancer. Here we identify and characterize SP cells from two distinct genetically engineered mouse ovarian cancer cell lines. Differential efflux of the DNA-binding dye Hoechst 33342 from these cell lines defined a human breast cancer-resistance protein 1-expressing, verapamil-sensitive SP of candidate cancer stem cells. In vivo, mouse SP cells formed measurable tumors sooner than non-SP (NSP) cells when equal numbers were injected into the dorsal fat pad of nude mice. The presence of Mullerian Inhibiting Substance (MIS) signaling pathway transduction molecules in both SP and NSP mouse cells led us to investigate the efficacy of MIS against these populations in comparison with traditional chemotherapies. MIS inhibited the proliferation of both SP and NSP cells, whereas the lipophilic chemotherapeutic agent doxorubicin more significantly inhibited the NSP cells. Finally, we identified breast cancer-resistance protein 1-expressing verapamil-sensitive SPs in three of four human ovarian cancer cell lines and four of six patient primary ascites cells. In the future, individualized therapy must incorporate analysis of the stem cell-like subpopulation of ovarian cancer cells when designing therapeutic strategies for ovarian cancer patients.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glicoproteínas/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Células-Tronco/citologia , Hormônios Testiculares/farmacologia , Animais , Hormônio Antimülleriano , Antineoplásicos/farmacologia , Benzimidazóis/farmacologia , Células CHO , Linhagem Celular Tumoral , Cricetinae , Feminino , Corantes Fluorescentes/farmacologia , Humanos , Camundongos , Transdução de Sinais , Verapamil/farmacologia
7.
Mol Cell Endocrinol ; 234(1-2): 87-93, 2005 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-15836957

RESUMO

Survival and growth of follicles in human ovarian tissue is presently only performed with limited success. We evaluated the effect of anti-Müllerian hormone (AMH) and/or testosterone on follicular growth during a 4-week culture period using ovarian cortical tissue from six women in their reproductive years. The cortex of each biopsy was isolated and immediately cryopreserved upon collection and stored in liquid nitrogen. After thawing the tissue was placed in culture. After the culture period all follicles were counted on histological sections and classified for viability and stage of development. Based on evaluation of 6603 follicles it was found that the number of growing follicles significantly increased during the culture period as compared to the uncultured control, irrespective of the composition of the culture medium. Furthermore, significantly more follicles advanced to the primary and secondary stage (p<0.05) in tissue cultured with AMH (54%) as compared to tissue cultured in control medium (41%). The mean diameter of follicles classified as primary follicles was significantly enhanced in tissue cultured in the presence of AMH (p=0.002) and AMH plus testosterone (p<0.001) as compared to that observed in tissue cultured with control medium and medium containing testosterone alone. In contrast the mean diameter of the oocyte and its nucleus remained similar irrespective of culture medium. In conclusion, AMH seems to affect early stages of human follicular development by enhancing recruitment, survival and/or growth during a 4-week culture period.


Assuntos
Glicoproteínas/farmacologia , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/crescimento & desenvolvimento , Hormônios Testiculares/farmacologia , Adulto , Hormônio Antimülleriano , Núcleo Celular/efeitos dos fármacos , Feminino , Glicoproteínas/fisiologia , Humanos , Folículo Ovariano/citologia , Hormônios Testiculares/fisiologia , Testosterona/farmacologia , Testosterona/fisiologia
8.
J Natl Cancer Inst Monogr ; (34): 18-21, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15784815

RESUMO

In this paper the role in the ovary of anti-Müllerian hormone (AMH), a member of the transforming growth factor-beta family of growth and differentiation factors, is reviewed. AMH has an inhibitory effect on primordial follicle recruitment and may also inhibit follicle-stimulating homone-dependent selection of follicles for dominance. In addition to its functional role in the ovary, AMH in serum is an excellent candidate marker as an indication of the ovarian reserve, not only in infertility clinic patients but also in women during and after cancer treatment.


Assuntos
Biomarcadores/análise , Glicoproteínas/farmacologia , Infertilidade Feminina/diagnóstico , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/fisiologia , Hormônios Testiculares/farmacologia , Hormônio Antimülleriano , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Diferenciação Celular , Feminino , Humanos , Ductos Paramesonéfricos , Neoplasias/tratamento farmacológico
9.
J Steroid Biochem Mol Biol ; 92(3): 199-208, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15555913

RESUMO

In addition to causing Müllerian duct regression in fetal males, Müllerian inhibiting substance (MIS) inhibits the expression of the bifunctional cytochrome P450, C17 hydroxylase/C(17-20) lyase (Cyp17), the enzyme that catalyzes the committed step in sex steroid synthesis. To investigate the paracrine effects of MIS on steroidogenic activity, we have performed assays with microsomes from mouse MA-10 Leydig cells. With microsomes from untreated MA-10 cells, progesterone was largely metabolized by 5alpha-reductase and subsequently converted by 3-keto steroid reductases to allopregnanolone and epiallopregnanolone. Addition of cAMP to the cells shifted microsomal steroid production to the Cyp17 product androstenedione and its 5alpha,3beta-reduced form, epiandrosterone. Microsomes from MIS-treated cells were less active with the progesterone substrate than those of untreated cells but co-treatment of the cells with both MIS and cAMP mitigated the cAMP-induced shift of the microsomes to androstenedione production. Quantitative analyses of steroid production by Cyp17 showed that cAMP decreased the amount of 17-hydroxyprogesterone produced relative to the androstenedione, suggesting that cAMP signaling lowers the efficiency of the Cyp17 hydroxylase activity or else increases the efficiency of its lyase activity. Addition of MIS to the cAMP-treated cells partially reversed this effect, as well. These results indicate that cAMP induces MA-10 cells to switch from producing 5alpha-reduced progesterone metabolites to producing androstenedione and its metabolites by increasing Cyp17 expression and its relative lyase activity, both of which are inhibited by MIS.


Assuntos
AMP Cíclico/farmacologia , Glicoproteínas/farmacologia , Células Intersticiais do Testículo/efeitos dos fármacos , Esteroides/biossíntese , Hormônios Testiculares/farmacologia , 17-alfa-Hidroxiprogesterona/metabolismo , Androstenodiona/biossíntese , Animais , Hormônio Antimülleriano , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos , Microssomos/efeitos dos fármacos , Microssomos/metabolismo , Progesterona/metabolismo , Ratos , Esteroide 17-alfa-Hidroxilase/efeitos dos fármacos
10.
Endocrinology ; 145(3): 1269-75, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14630719

RESUMO

Müllerian-inhibiting substance (MIS) reduces testosterone synthesis in Leydig cells by inhibiting cytochrome P450C17 hydroxylase/C17-20 lyase expression. However, in mouse Leydig MA-10 cells, MIS also enhances the cAMP-induced expression of mRNA for steroidogenic acute regulatory protein (StAR), which transports cholesterol to the inner mitochondrial membrane for conversion to pregnenolone. We hypothesized that the MIS-induced StAR expression is the indirect result of reduced testosterone synthesis in Leydig cells caused by MIS. We show that, in addition to MIS, flutamide, an androgen receptor antagonist, enhanced StAR mRNA expression when added to cAMP-treated MA-10 cells, whereas dihydrotestosterone, a potent androgen receptor agonist, attenuated these responses. Progesterone, dexamethasone, and estradiol also inhibited StAR mRNA expression. Addition of MIS to cAMP-treated MA-10 cells transfected with a StAR-promoter luciferase reporter resulted in increased StAR promoter activity over cAMP alone; this effect was inhibited by dihydrotestosterone, suggesting that androgens inhibit StAR mRNA expression at the transcriptional level. Androgen-mediated inhibition of StAR expression was also observed in primary Leydig cell culture and in vivo using both hypophysectomized mice and mice treated with the GnRH antagonist, acyline. These results suggest that the induction of StAR expression by MIS occurs secondary to the MIS-mediated reduction in testosterone synthesis by relieving a hitherto uncharacterized androgen-dependent feedback inhibition on StAR expression. These findings may impact future treatment strategies aimed at reducing androgen; for example, in the treatment of prostatic cancer, antiandrogen treatment might benefit from adjuvant therapy to inhibit StAR expression.


Assuntos
Androgênios/farmacologia , Di-Hidrotestosterona/farmacologia , Glicoproteínas/farmacologia , Fosfoproteínas/genética , Hormônios Testiculares/farmacologia , Testosterona/biossíntese , Antagonistas de Androgênios/farmacologia , Animais , Hormônio Antimülleriano , Linhagem Celular Tumoral , Retroalimentação Fisiológica/efeitos dos fármacos , Flutamida/farmacologia , Expressão Gênica/efeitos dos fármacos , Hipofisectomia , Técnicas In Vitro , Tumor de Células de Leydig , Masculino , Camundongos , Camundongos Endogâmicos , RNA Mensageiro/análise , Transcrição Gênica/efeitos dos fármacos
11.
Biol Reprod ; 70(3): 600-7, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14585814

RESUMO

The postnatal development of Leydig cell precursors is postulated to be controlled by Sertoli cell secreted factors, which may have a determinative influence on Leydig cell number and function in sexually mature animals. One such hormone, Mullerian inhibiting substance (MIS), has been shown to inhibit DNA synthesis and steroidogenesis in primary Leydig cells and Leydig cell tumor lines. To further delineate the effects of MIS on Leydig cell proliferation and steroidogenesis, we employed the established ethylene dimethanesulphonate (EDS) model of Leydig cell regeneration. Following EDS ablation of differentiated Leydig cells in young adult rats, recombinant MIS or vehicle was delivered by intratesticular injection for 4 days (Days 11-14 after EDS). On Days 15 and 35 after EDS (1 and 21 days post-MIS injections), endocrine function was assessed and testes were collected for stereology, immunohistochemistry, and assessment of proliferation and steroidogenesis. Although serum testosterone and luteinizing hormone (LH) were no different, intratesticular testosterone was higher on Day 35 in MIS-treated animals. At both time points, intratesticular 5alpha-androstan-3alpha,17beta-diol concentrations were much higher than that of testosterone. MIS-treated animals had fewer mesenchymal precursors on Day 15 and fewer differentiated Leydig cells on Day 35 with decreased numbers of BrdU+ nuclei. Apoptotic interstitial cells were observed only in the MIS-treated testes, not in the vehicle-treated group on Day 15. These data suggest that MIS inhibits regeneration of Leydig cells in EDS-treated rats by enhancing apoptotic cell death as well as by decreasing proliferative capacity.


Assuntos
Glicoproteínas/farmacologia , Células Intersticiais do Testículo/citologia , Células Intersticiais do Testículo/efeitos dos fármacos , Hormônios Testiculares/farmacologia , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase/metabolismo , Animais , Hormônio Antimülleriano , Contagem de Células , Divisão Celular/efeitos dos fármacos , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Marcação In Situ das Extremidades Cortadas , Hormônio Luteinizante/sangue , Masculino , Mesilatos , Tamanho do Órgão , Ratos , Ratos Sprague-Dawley , Esteroide 17-alfa-Hidroxilase/metabolismo , Testículo/citologia , Testosterona/sangue
12.
Mol Cell Endocrinol ; 211(1-2): 1-7, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14656469

RESUMO

Migration of mesonephric cells into XY gonads is a critical early event in testis cord formation. Based on the fact that anti-Müllerian hormone (AMH) can induce testis cord formation in XX gonads, we investigated whether AMH plays a role in the induction of cell migration. Addition of recombinant AMH induced mesonephric migration into XX gonads in culture. AMH-treated XX gonads displayed increased vascular development and altered morphology of the coelomic epithelium, both features of normal testis differentiation. AMH did not induce markers of Sertoli or Leydig cell differentiation. We examined early testis development in Amh-deficient mice, but found no abnormalities, suggesting that any function AMH may have in vivo is redundant. Other transforming growth factor (TGF-beta) family proteins, bone morphogenetic proteins (BMP2 and BMP4) show similar inductive effects on XX gonads in culture. Although neither BMP2 nor BMP4 is expressed in embryonic XY gonads, our findings suggest that a TGF-beta signalling pathway endogenous to the XY gonad may be involved in regulation of mesonephric cell migration. The factors involved in this process remain to be identified.


Assuntos
Movimento Celular/fisiologia , Glicoproteínas/fisiologia , Gônadas/embriologia , Mesonefro/citologia , Hormônios Testiculares/fisiologia , Fator de Crescimento Transformador beta , Animais , Hormônio Antimülleriano , Proteína Morfogenética Óssea 2 , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/farmacologia , Movimento Celular/efeitos dos fármacos , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Técnicas de Cocultura , Epitélio/anatomia & histologia , Epitélio/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/genética , Glicoproteínas/farmacologia , Gônadas/efeitos dos fármacos , Gônadas/metabolismo , Proteínas de Grupo de Alta Mobilidade/genética , Humanos , Imuno-Histoquímica , Hibridização In Situ , Óperon Lac/genética , Laminina/análise , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neovascularização Fisiológica/fisiologia , Técnicas de Cultura de Órgãos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta , Proteínas Recombinantes/farmacologia , Fatores de Transcrição SOX9 , Diferenciação Sexual/fisiologia , Hormônios Testiculares/genética , Hormônios Testiculares/farmacologia , Fatores de Transcrição/genética , beta-Galactosidase/análise
13.
Mol Cell Endocrinol ; 211(1-2): 37-42, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14656474

RESUMO

It is almost 60 years since Prof. Alfred Jost reported the seminal observations regarding Müllerian inhibiting substance (MIS). His experiments clearly showed that a testicular product other than testosterone, a Müllerian inhibitor, was responsible for Müllerian duct regression. Twenty-five years later Dr. Picon established an organ culture assay which paved the way for the initial studies into the biochemistry and biology of Müllerian inhibiting substance, also known as Anti-Müllerian hormone (AMH), undertaken first in Dr. Nathalie Josso's Laboratory in Paris then in our own laboratory in Boston. Purification of MIS led to cloning the human gene and production of recombinant human (rhMIS). MIS is a 140 kDa glycoprotein homodimer which is activated by a biosynthetic protease, cleaving MIS into an aminoterminus (110 kDa) and a carboxyterminus (25 kDa). The latter domain is sufficient for biological activities. MIS functions by interacting with two receptors; a type II binds the hormone and at type I that initiates downstream signaling. The MIS type II receptor has been cloned and functionally confirmed as distinct from that of other members of the TGFbeta superfamily. MIS can employ a number of type I receptors (ALK2, ALK3, ALK6) and BMP receptor specific SMADS 1, 5, and 8 in various tissue specific contexts. Cell lines derived from human ovarian, breast, and prostate tumors, and from rodent Leydig cell tumors, which respond to MIS in growth inhibition assays, all express the MIS type II receptor. A variety of signal transduction pathways are associated with the grown inhibition mediated by MIS. For example, breast and prostate cancer cell lines use a MIS-mediated NFkappaB pathway leading to G1 arrest and apoptosis. The ovarian cancer cell lines employ a pathway which enhances p16, modulates the E2Fs, and induces apoptosis. These signal transduction events can establish new rational treatment strategies to complement the growth inhibitory effects mediated by MIS. These combination strategies are being tested in vitro, and where appropriate will be tested in vivo using the highly purified MIS preparations, prior to use in early human clinical trials.


Assuntos
Glicoproteínas/biossíntese , Glicoproteínas/uso terapêutico , Hormônios Testiculares/biossíntese , Hormônios Testiculares/uso terapêutico , Receptores de Ativinas Tipo I/fisiologia , Animais , Hormônio Antimülleriano , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Feminino , Fibrinolisina/metabolismo , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/farmacologia , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Receptores de Peptídeos/fisiologia , Receptores de Fatores de Crescimento Transformadores beta , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Diferenciação Sexual/fisiologia , Hormônios Testiculares/farmacologia
14.
Mol Cell Endocrinol ; 211(1-2): 43-9, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14656475

RESUMO

The MIS type II receptor is expressed at high levels in the Mullerian duct and in Sertoli cells and granulosa cells of the embryonic and adult gonads. The presence of MIS type II and type I receptors in tissues and cell lines derived from breast and prostate suggests that the prostate and mammary glands may be additional targets for MIS action. In both breast and prostate cancer cells, MIS activated NFkB DNA binding activity and induced IEX-1, an immediate early gene which regulates cell growth and apoptosis. Exposure of cells to MIS inhibited growth by increasing the fraction of cells in the G1 phase of the cell cycle and by inducing apoptosis. These results suggest that MIS may be a putative mediator of growth regulatory signals in the breast and prostate.


Assuntos
Glicoproteínas/farmacologia , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Hormônios Testiculares/farmacologia , Receptores de Ativinas Tipo I/genética , Animais , Hormônio Antimülleriano , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Imediatamente Precoces/genética , Masculino , Proteínas de Membrana , NF-kappa B/genética , Subunidade p50 de NF-kappa B , Proteínas de Neoplasias/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento/genética , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta , Proteínas Recombinantes/farmacologia , Fator de Transcrição RelA
15.
Mol Cell Endocrinol ; 211(1-2): 99-104, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14656482

RESUMO

Müllerian-inhibiting substance (MIS), a member of the transforming growth factor-beta family of cytokines that signal through a heteromeric complex of single-transmembrane serine/threonine kinase receptors, is required for Müllerian duct regression and normal reproductive tract development in the male embryo. However, the continued expression of MIS at high levels in males until puberty and its induction in females after birth suggested other roles for MIS. Additionally, Leydig cell development and steroidogenic capacity and ovarian follicle recruitment were abnormal in MIS-knockout or MIS-overexpressing mice. We have shown that MIS inhibits the cAMP-induced expression of cytochrome P450 C17alpha-hydroxylase/C17-20 lyase (Cyp17) mRNA both in vitro and in vivo. Our current efforts are to understand the molecular mechanisms regulating both MIS type II receptor (MISRII) expression and its signaling in rodent Leydig cell lines. MISRII expression in R2C cells requires both steroidogenic factor-1 and an unknown protein to bind to its proximal promoter in the context of 1.6 kb 5'-flanking DNA. When bound by MIS, signaling by the receptor in MA-10 cells blocks the protein kinase A-mediated induction of Cyp17 expression by a cAMP regulatory element-binding protein independent mechanism. We continue to investigate the molecular mechanisms of MISRII expression and possible interactions between MIS-regulated SMAD activation and cAMP signaling. These studies will provide a better understanding of the role played by MIS during postnatal life.


Assuntos
Glicoproteínas/fisiologia , Células Intersticiais do Testículo/metabolismo , Esteroides/biossíntese , Hormônios Testiculares/fisiologia , 17-alfa-Hidroxiprogesterona/sangue , 17-alfa-Hidroxiprogesterona/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Hormônio Antimülleriano , Linhagem Celular Tumoral , Gonadotropina Coriônica/farmacologia , AMP Cíclico/farmacologia , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Fatores de Transcrição Fushi Tarazu , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/farmacologia , Células Intersticiais do Testículo/efeitos dos fármacos , Masculino , Camundongos , Progesterona/sangue , Progesterona/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Ratos , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta , Transdução de Sinais/fisiologia , Esteroide 17-alfa-Hidroxilase/genética , Fator Esteroidogênico 1 , Hormônios Testiculares/farmacologia , Testosterona/sangue , Testosterona/metabolismo , Fatores de Transcrição/metabolismo
16.
J Biol Chem ; 278(51): 51703-12, 2003 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-14532292

RESUMO

This report demonstrates that in addition to interferons and cytokines, members of the TGF beta superfamily such as Mullerian inhibiting substance (MIS) and activin A also regulate IRF-1 expression. MIS induced IRF-1 expression in the mammary glands of mice in vivo and in breast cancer cells in vitro and stimulation of IRF-1 by MIS was dependent on activation of the NF kappa B pathway. In the rat mammary gland, IRF-1 expression gradually decreased during pregnancy and lactation but increased at involution. In breast cancer, the IRF-1 protein was absent in 13% of tumors tested compared with matched normal glands. Consistent with its growth suppressive activity, expression of IRF-1 in breast cancer cells induced apoptosis. Treatment of breast cancer cells with MIS and interferon gamma (IFN-gamma) co-stimulated IRF-1 and CEACAM1 expression and synergistic induction of CEACAM1 by a combination of MIS and IFN-gamma was impaired by antisense IRF-1 expression. Furthermore, a combination of IFN-gamma and MIS inhibited the growth of breast cancer cells to a greater extent than either one alone. Both reagents alone significantly decreased the fraction of cells in the S-phase of the cell cycle, an effect not enhanced when they were used in combination. However, MIS promoted IFN-gamma-induced apoptosis demonstrating a functional interaction between these two classes of signaling molecules in regulation of breast cancer cell growth.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/farmacologia , Interferon gama/farmacologia , Hormônios Testiculares/farmacologia , Ativinas/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Hormônio Antimülleriano , Biópsia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/biossíntese , Sinergismo Farmacológico , Feminino , Humanos , Subunidades beta de Inibinas/farmacologia , Fator Regulador 1 de Interferon , Camundongos , Camundongos Endogâmicos C3H , Pessoa de Meia-Idade , Fosfoproteínas/análise , Fosfoproteínas/biossíntese , Ratos , Fase S/efeitos dos fármacos
17.
Reproduction ; 124(5): 601-9, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12416998

RESUMO

Anti-Müllerian hormone (AMH), also known as Müllerian inhibiting substance, is a member of the transforming growth factor beta superfamily of growth and differentiation factors. In contrast to other members of the family, which exert a broad range of functions in multiple tissues, the principal function of AMH is to induce regression of the Müllerian ducts during male sex differentiation. However, the patterns of expression of AMH and its type II receptor in the postnatal ovary indicate that AMH may play an important role in ovarian folliculogenesis. This review describes several in vivo and in vitro studies showing that AMH participates in two critical selection points of follicle development: it inhibits the recruitment of primordial follicles into the pool of growing follicles and also decreases the responsiveness of growing follicles to FSH.


Assuntos
Glicoproteínas , Inibidores do Crescimento/fisiologia , Ductos Paramesonéfricos/embriologia , Ovário/fisiologia , Transdução de Sinais/fisiologia , Hormônios Testiculares/fisiologia , Animais , Hormônio Antimülleriano , Biomarcadores/sangue , Biomarcadores Tumorais/sangue , Depressão Química , Feminino , Hormônio Foliculoestimulante/farmacologia , Expressão Gênica , Tumor de Células da Granulosa/diagnóstico , Células da Granulosa/metabolismo , Inibidores do Crescimento/farmacologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia , Neoplasias Ovarianas/diagnóstico , Ratos , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta , Hormônios Testiculares/farmacologia
18.
Endocrinology ; 143(9): 3351-60, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12193547

RESUMO

Müllerian inhibiting substance (MIS) is produced by fetal Sertoli cells and causes regression of the Müllerian duct in male fetuses shortly after commitment of the bipotential embryonic gonad to testes differentiation. MIS is also produced by the Sertoli cells and granulosa cells of the adult gonads where it plays an important role in regulating steroidogenesis. We have previously shown that MIS can dramatically reduce testosterone synthesis in Leydig cells by inhibiting the expression of cytochrome P450 17alpha-hydroxylase/C(17-20) lyase (Cyp17) mRNA in vitro and in vivo. To characterize the signal transduction pathway used by MIS to control expression of endogenous Cyp17 in a mouse Leydig cell line, we demonstrate that MIS inhibits both LH- and cAMP-induced expression of Cyp17 at concentrations as low as 3.5 nM and for as long as 18 h. The induction of steroidogenic acute regulatory protein (StAR) mRNA by cAMP, however, was slightly increased by addition of MIS. Protein kinase A (PKA) inhibition with H-89 blocked Cyp17 mRNA induction, suggesting that MIS interferes with the PKA signal transduction pathway. Inhibition of Cyp17 induction was not seen with added U0126, and wortmannin inhibited the induction incompletely. In addition, phosphorylation of the cAMP responsive element binding protein (CREB) was not detected following 50 micro M cAMP exposure, a concentration sufficient for Cyp17 mRNA induction. Moreover, CREB phosphorylation, which was observed with addition of 500 micro M cAMP, was not inhibited by coincubation with MIS. Taken together, these results suggest that cAMP induces expression of Cyp17 by a PKA-mediated mechanism and that this induction, which is inhibited by MIS signal transduction, does not require CREB activity, and is distinct from that used to induce steroidogenic acute regulatory protein expression.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Expressão Gênica/efeitos dos fármacos , Glicoproteínas , Inibidores do Crescimento/farmacologia , Células Intersticiais do Testículo/enzimologia , Esteroide 17-alfa-Hidroxilase/genética , Hormônios Testiculares/farmacologia , Animais , Hormônio Antimülleriano , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Relação Dose-Resposta a Droga , Inibidores do Crescimento/administração & dosagem , Cinética , Tumor de Células de Leydig , Masculino , Camundongos , Fosforilação , RNA Mensageiro/análise , Transdução de Sinais , Hormônios Testiculares/administração & dosagem , Células Tumorais Cultivadas
19.
J Biol Chem ; 275(47): 37101-9, 2000 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-10958795

RESUMO

Müllerian inhibiting substance (MIS), a transforming growth factor-beta family member, causes regression of the Müllerian duct in male embryos. MIS overexpression in transgenic mice ablates the ovary, and MIS inhibits the growth of ovarian cancer cell lines in vitro, suggesting a key role for this hormone in postnatal development of the ovary. This report describes a mechanism for MIS-mediated growth inhibition in both a human epithelial ovarian cancer cell line and a cell line derived from normal ovarian surface epithelium, which is the origin of human epithelial ovarian cancers. MIS-treated cells accumulated in the G(1) phase of the cell cycle and subsequently underwent apoptosis. MIS up-regulated the cyclin-dependent kinase inhibitor p16 through an MIS type II receptor-mediated mechanism and inhibited growth in the absence of detectable or inactive Rb protein. Prolonged treatment with MIS down-regulated the Rb-related protein p130 and increased the Rb family-regulated transcription factor E2F1, overexpression of which inhibited growth. These findings demonstrate that p16 is required for MIS-mediated growth inhibition in ovarian epithelial cells and tumor cells and suggest that up-regulation of E2F1 also plays a role in this process.


Assuntos
Glicoproteínas , Inibidores do Crescimento/farmacologia , Ovário/patologia , Proteínas , Rubídio/metabolismo , Hormônios Testiculares/farmacologia , Animais , Hormônio Antimülleriano , Proteínas Sanguíneas/metabolismo , Proteínas de Ligação a Calmodulina/metabolismo , Diferenciação Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Feminino , Humanos , Ligantes , Masculino , Camundongos , Neoplasias Ovarianas/patologia , Ovário/efeitos dos fármacos , Proteína p130 Retinoblastoma-Like , Células Tumorais Cultivadas
20.
J Biol Chem ; 275(36): 27973-8, 2000 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-10854429

RESUMO

Anti-Müllerian hormone induces the regression of fetal Müllerian ducts and inhibits the transcription of gonadal steroidogenic enzymes. It belongs to the transforming growth factor-beta family whose members signal through a pair of serine/threonine kinase receptors and Smad effectors. Only the anti-Müllerian hormone type II receptor has been identified. Our goal was to determine whether anti-Müllerian hormone could share a type I receptor with another family member. Co-immunoprecipitation of known type I receptors with anti-Müllerian hormone type II receptor clearly showed that the bone morphogenetic protein type IB receptor was the only cloned type I receptor interacting in a ligand-dependent manner with this type II receptor. Anti-Müllerian hormone also activates the bone morphogenetic protein-specific Smad1 pathway and the XVent2 reporter gene, an anti-Müllerian hormone type II receptor-dependent effect abrogated by a dominant negative version of bone morphogenetic protein type IB receptor. Reverse amplification experiments showed that bone morphogenetic protein type IB receptor is co-expressed with anti-Müllerian hormone type II receptor in most anti-Müllerian hormone target tissues. Our data support a model in which a ligand, anti-Müllerian hormone, gains access to a shared type I receptor and Smad1 system through a highly restricted type II receptor.


Assuntos
Proteínas de Ligação a DNA , Glicoproteínas , Inibidores do Crescimento/farmacologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas/metabolismo , Receptores de Fatores de Crescimento/fisiologia , Receptores de Peptídeos/fisiologia , Hormônios Testiculares/farmacologia , Transativadores , Animais , Hormônio Antimülleriano , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Receptores de Proteínas Morfogenéticas Ósseas Tipo II , Células CHO , Linhagem Celular , Cricetinae , Genes Reporter , Humanos , Camundongos , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Receptores de Superfície Celular/fisiologia , Receptores de Fatores de Crescimento/efeitos dos fármacos , Receptores de Fatores de Crescimento/genética , Receptores de Peptídeos/efeitos dos fármacos , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad , Proteína Smad1 , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA