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











Base de dados
Intervalo de ano de publicação
1.
Reprod Domest Anim ; 52 Suppl 2: 354-358, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27859771

RESUMO

The intent of this contribution is to provide an update of the progress we have made towards developing a method/treatment to permanently sterilize cats. Our approach employs two complementary methodologies: RNA interference (RNAi) to silence genes involved in the central control of reproduction and a virus-based gene therapy system intended to deliver RNAi selectively to the hypothalamus (where these genes are expressed) via the systemic administration of modified viruses. We selected the hypothalamus because it contains neurons expressing Kiss1 and Tac3, two genes essential for reproduction and fertility. We chose the non-pathogenic adeno-associated virus (AAV) as a vector whose tropism could be modified to target the hypothalamus. The issues that must be overcome to utilize this vector as a delivery vehicle to induce sterility include modification of the wild-type AAV to target the hypothalamic region of the brain with a simultaneous reduction in targeting of peripheral tissues and non-hypothalamic brain regions, identification of RNAi targets that will effectively reduce the expression of Kiss1 and Tac3 without off-target effects, and determination if neutralizing antibodies to the AAV serotype of choice are present in cats. Successful resolution of these issues will pave the way for the development of a powerful tool to induce the permanent sterility in cats.


Assuntos
Gatos , Anticoncepção/veterinária , Dependovirus , Inativação Gênica , Vetores Genéticos , Hipotálamo , Animais , Anticoncepção/métodos , Expressão Gênica/efeitos dos fármacos , Engenharia Genética/métodos , Engenharia Genética/veterinária , Infertilidade/etiologia , Infertilidade/veterinária , Kisspeptinas/antagonistas & inibidores , Kisspeptinas/genética , Neurocinina B/antagonistas & inibidores , Neurocinina B/genética , Interferência de RNA
2.
Endocrinology ; 154(2): 942-55, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23291449

RESUMO

Lin28 and Lin28b are related RNA-binding proteins that inhibit the maturation of miRNAs of the let-7 family and participate in the control of cellular stemness and early embryonic development. Considerable interest has arisen recently concerning other physiological roles of the Lin28/let-7 axis, including its potential involvement in the control of puberty, as suggested by genome-wide association studies and functional genomics. We report herein the expression profiles of Lin28 and let-7 members in the rat hypothalamus during postnatal maturation and in selected models of altered puberty. The expression patterns of c-Myc (upstream positive regulator of Lin28), mir-145 (negative regulator of c-Myc), and mir-132 and mir-9 (putative miRNA repressors of Lin28, predicted by bioinformatic algorithms) were also explored. In male and female rats, Lin28, Lin28b, and c-Myc mRNAs displayed very high hypothalamic expression during the neonatal period, markedly decreased during the infantile-to-juvenile transition and reached minimal levels before/around puberty. A similar puberty-related decline was observed for Lin28b in monkey hypothalamus but not in the rat cortex, suggesting species conservation and tissue specificity. Conversely, let-7a, let-7b, mir-132, and mir-145, but not mir-9, showed opposite expression profiles. Perturbation of brain sex differentiation and puberty, by neonatal treatment with estrogen or androgen, altered the expression ratios of Lin28/let-7 at the time of puberty. Changes in the c-Myc/Lin28b/let-7 pathway were also detected in models of delayed puberty linked to early photoperiod manipulation and, to a lesser extent, postnatal underfeeding or chronic subnutrition. Altogether, our data are the first to document dramatic changes in the expression of the Lin28/let-7 axis in the rat hypothalamus during the postnatal maturation and after different manipulations that disturb puberty, thus suggesting the potential involvement of developmental changes in hypothalamic Lin28/let-7 expression in the mechanisms permitting/leading to puberty onset.


Assuntos
Envelhecimento/genética , Encéfalo/crescimento & desenvolvimento , MicroRNAs/metabolismo , Proteínas de Ligação a RNA/biossíntese , Animais , Células-Tronco Embrionárias/citologia , Feminino , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Masculino , MicroRNAs/biossíntese , Proteínas Proto-Oncogênicas c-myc/biossíntese , Puberdade/efeitos dos fármacos , Ratos , Ratos Wistar , Distribuição Tecidual
3.
Hum Reprod ; 27(11): 3249-58, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22888166

RESUMO

STUDY QUESTION: Is decorin (DCN), a putative modulator of growth factor (GF) signaling, expressed in the primate ovary and does it play a role in ovarian biology? SUMMARY ANSWER: DCN expression in the theca, the corpus luteum (CL), its presence in the follicular fluid (FF) and its actions revealed in human IVF-derived granulosa cells (GCs), suggest that it plays multiple roles in the ovary including folliculogenesis, ovulation and survival of the CL. WHAT IS KNOWN ALREADY: DCN is a secreted proteoglycan, which has a structural role in the extracellular matrix (ECM) and also interferes with the signaling of multiple GF/GF receptors (GFRs). However, DCN expression and action in the primate ovary has yet to be determined. STUDY DESIGN, SIZE, DURATION: Archival human and monkey ovarian samples were analyzed. Studies were conducted using FF and GC samples collected from IVF patients. PARTICIPANTS/MATERIALS, SETTING, METHODS: Immunohistochemistry, western blotting, RT-PCR, quantitative RT-PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA) studies were complemented by cellular studies, including the measurements of intracellular Ca²âº, reactive oxygen species (ROS), epidermal GF receptor (EGFR) phosphorylation by DCN and caspase activity. MAIN RESULTS AND THE ROLE OF CHANCE: Immunohistochemistry revealed strong DCN staining in the connective tissue and follicular thecal compartments, but not in GCs of pre-antral and antral follicles. Pre-ovulatory follicles could not be studied, but DCN was associated with connective tissue of CL samples and the cytoplasm of luteal cells. DCN expression in monkey CL doubled (P < 0.05) towards the end of the luteal lifespan. DCN was found in human FF obtained from IVF patients (mean: 12.9 ng/ml; n = 20) as determined by ELISA. DCN mRNA and/or protein were detected in freshly isolated and cultured, luteinized human GCs. In the latter, exogenous human recombinant DCN increased intracellular Ca²âº levels and induced the production of ROS in a concentration-dependent manner. DCN, like epidermal GF, phosphorylated EGFR significantly (P < 0.05) and reduced the activity of caspase 3/7 in cultured GCs. The data indicate the expression of DCN in the theca of growing follicles, in FF of ovulatory follicles and in the CL. Therefore, DCN may exert paracrine actions via GF/GFR systems in multiple ovarian compartments. LIMITATIONS, REASONS FOR CAUTION: Functional studies were performed in cultures of human luteinized GCs, which are an apt model but may not fully mirror the pre-ovulatory GC compartment or the CL. Other human ovarian cells, including the thecal cells, were not available. WIDER IMPLICATIONS OF THE FINDINGS: In accordance with its evolving roles in other organs, ovarian DCN is an ECM-associated component, which acts as a multifunctional regulator of GF signaling in the primate ovary. DCN may thus be involved in folliculogenesis, ovulation and the regulation of the CL survival in primates. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by Deutsche Forschungsgemeinschaft (DFG) MA1080/17-3 and in part DFG MA1080/21-1 (to AM), NIH grants HD24870 (S.R.O. and R.L.S.), the Eunice Kennedy Shriver NICHD/NIH through cooperative agreement HD18185 as part of the Specialized Cooperative Centers Program in Reproduction and Infertility Research (S.R.O.) and 8P51OD011092-53 for the operation of the Oregon National Primate Research Center (G.A.D., J.D.H., S.R.O. and R.L.S).


Assuntos
Decorina/metabolismo , Matriz Extracelular/metabolismo , Fase Luteal/metabolismo , Oogênese , Ovário/metabolismo , Ovulação/metabolismo , Adulto , Animais , Células Cultivadas , Corpo Lúteo/citologia , Corpo Lúteo/metabolismo , Decorina/genética , Receptores ErbB/metabolismo , Feminino , Líquido Folicular/metabolismo , Regulação da Expressão Gênica , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Humanos , Macaca mulatta , Ovário/citologia , Fosforilação , Processamento de Proteína Pós-Traducional , RNA Mensageiro/metabolismo , Células Tecais/citologia , Células Tecais/metabolismo
4.
Reprod Domest Anim ; 47 Suppl 4: 228-32, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22827375

RESUMO

A non-surgical method to induce sterility would be a useful tool to control feral populations of animals. Our laboratories have experience with approaches aimed at targeting brain cells in vivo with vehicles that deliver a payload of either inhibitory RNAs or genes intended to correct cellular dysfunction. A combination/modification of these methods may provide a useful framework for the design of approaches that can be used to sterilize cats and dogs. For this approach to succeed, it has to meet several conditions: it needs to target a gene essential for fertility. It must involve a method that can selectively silence the gene of interest. It also needs to deliver the silencing agent via a minimally invasive method. Finally, the silencing effect needs to be sustained for many years, so that expansion of the targeted population can be effectively prevented. In this article, we discuss this subject and provide a succinct account of our previous experience with: (i) molecular reagents able to disrupt reproductive cyclicity when delivered to regions of the brain involved in the control of reproduction and (ii) molecular reagents able to ameliorate neuronal disease when delivered systemically using a novel approach of gene therapy.


Assuntos
Interferência de RNA/fisiologia , Esterilização Reprodutiva/veterinária , Adenoviridae , Animais , Gatos , Cães , Feminino , Fertilidade/fisiologia , Vetores Genéticos , Hipotálamo/fisiologia , Infertilidade Feminina , Masculino , MicroRNAs , Controle da População , Primatas , Ratos , Esterilização Reprodutiva/métodos
5.
Hum Reprod ; 25(4): 969-76, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20097922

RESUMO

BACKGROUND: Oxytocin (OT) is produced by granulosa cells (GCs) of pre-ovulatory ovarian follicles and the corpus luteum (CL) in some mammalian species. Actions of OT in the ovary have been linked to luteinization, steroidogenesis and luteolysis. Human IVF-derived (h)GCs possess a functional OT receptor (OTR), linked to elevation of intracellular Ca(2+), but molecular identity of the receptor for OT in human granulosa cells (hGCs) and down-stream consequences are not known. METHODS AND RESULTS: RT-PCR, sequencing and immunocytochemistry identified the genuine OTR in hGCs. OT (10 nM-10 microM) induced elevations of intracellular Ca(2+) levels (Fluo-4 measurements), which were blocked by tocinoic acid (TA; 50 microM, a selective OTR-antagonist). Down-stream effects of OTR-activation include a concentration dependent decrease in cell viability/metabolism, manifested by reduced ATP-levels, increased caspase3/7-activity (P < 0.05) and electron microscopical signs of cellular regression. TA blocked all of these changes. Immunoreactive OTR was found in the CL and GCs of large and, surprisingly, also small pre-antral follicles of the human ovary. Immunoreactive OTR in the rhesus monkey ovary was detected in primordial and growing primary follicles in the infantile ovary and in follicles at all stages of development in the adult ovary, as well as the CL: these results were corroborated by RT-PCR analysis of GCs excised by laser capture microdissection. CONCLUSIONS: Our study identifies genuine OTRs in human and rhesus monkey GCs. Activation by high levels of OT leads to cellular regression in hGCs. As GCs of small follicles also express OTRs, OT may have as yet unknown functions in follicular development.


Assuntos
Apoptose/genética , Apoptose/fisiologia , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Macaca mulatta/genética , Macaca mulatta/metabolismo , Ovário/citologia , Ovário/metabolismo , Receptores de Ocitocina/genética , Receptores de Ocitocina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Sequência de Bases , Sinalização do Cálcio , Corpo Lúteo/citologia , Corpo Lúteo/metabolismo , Primers do DNA/genética , Feminino , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Microscopia Eletrônica de Transmissão , Ocitocina/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie
6.
Endocrinology ; 142(5): 2078-86, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11316775

RESUMO

Nerve growth factor (NGF) epitomizes a family of proteins known as the neurotrophins (NTs), which are required for the survival and differentiation of neurons within both the central and peripheral nervous system. Synthesis of NGF in tissues innervated by the peripheral nervous system is consistent with its function as a target-derived trophic factor. However, the presence of low- and high-affinity NGF receptors in the gonads suggests another function for the NTs within the reproductive endocrine system. We now report that NGF is required for the growth of primordial ovarian follicles, a process known to occur independently of pituitary gonadotropins. Both the NT receptor p75(NTR) and the NGF tyrosine kinase receptor trkA were found to be expressed in the ovaries of infantile normal mice and mice carrying a null mutation of the NGF gene. The ovaries from homozygote NGF-null (-/-) mutant animals, analyzed after completion of ovarian histogenesis, exhibited a markedly reduced population of primary and secondary follicles in the presence of normal serum gonadotropin levels, and an increased number of oocytes that failed to be incorporated into a follicular structure. Assessment of mitogenic activity using two complementary proliferation markers revealed a conspicuous reduction in somatic cell proliferation in the ovaries of NGF-deficient mice. These results suggest that the delay in follicular growth observed in NGF(-/-) mice may be related to the loss of a proliferative signal provided by NGF to the nonneural endocrine component of the ovary.


Assuntos
Fator de Crescimento Neural/fisiologia , Folículo Ovariano/fisiologia , Receptor trkA/genética , Animais , Divisão Celular , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Neural/genética , Ovário/patologia , Antígeno Nuclear de Célula em Proliferação/análise , Receptor de Fator de Crescimento Neural , Receptor trkA/análise , Receptores de Fator de Crescimento Neural/análise
7.
Endocrinology ; 141(12): 4736-50, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11108289

RESUMO

TrkA, the nerve growth factor (NGF) tyrosine kinase receptor, is expressed not only in the nervous system, but also in nonneural cells, including discrete cellular subsets of the endocrine and immune system. In the rat ovary, trkA receptor abundance increases strikingly in thecal-interstitial cells during the hours preceding the first ovulation. Blockade of either trkA transducing capacity or NGF biological activity inhibited ovulation, suggesting a role for NGF in the ovulatory process of this species. To identify some of the processes that may be affected by trkA activation in the thecal compartment, we used purified thecal cells/thecal fibroblasts from bovine ovaries (heretofore referred to as thecal cells). Ribonuclease protection assays employing bovine-specific cRNA probes demonstrated the presence of the messenger RNAs (mRNAs) encoding NGF and its receptors, p75 NTR and trkA, in the thecal compartment of small, medium, and large antral follicles and showed that trkA mRNA is also expressed in granulosa cells. In situ hybridization and immunohistochemical examination of intact ovaries confirmed these cellular sites of NGF and trkA synthesis. TrkA mRNA, but not NGF mRNA, was lost within 48 h of placing thecal cells in culture. Thus, to study trkA-mediated actions of NGF on these cells we transiently expressed the receptor by transfection with a vector containing a full-length rat trkA complementary DNA under transcriptional control of the cytomegalovirus promoter. Because ovulation is preceded by an LH-dependent increase in androgen and progesterone production, the ability of NGF to modify the release of these steroids was determined in freshly plated cells still containing endogenous trkA receptors and in cells undergoing luteinization in culture that were transiently transfected with the trkA-encoding plasmid. NGF stimulated both androgen and progesterone release in freshly plated thecal cells, but not in luteinizing cells provided with trkA receptors. As ovulation in rodents requires an increased formation of PGE2 and has been shown to be antedated by proliferation of thecal fibroblasts, we determined the ability of NGF to affect these parameters in trkA-transfected thecal cells. The neurotrophin rapidly stimulated PGE2 release and amplified the early steroidal response to hCG in trkA-expressing cells, but not in cells lacking the receptor. Likewise, NGF stimulated [3H]thymidine incorporation into trkA-containing cells, but not into cells that had lost the receptor in culture. Induction of ovulation in immature rats by gonadotropin treatment verified that an increased cell proliferation in the thecal compartment, determined by the incorporation of bromodeoxyuridine into cell nuclei, occurs 4-5 h before ovulation in this species. These results suggest that the contribution of NGF to the ovulatory process includes a stimulatory effect of the neurotrophin on steroidogenesis, PGE2 formation, and proliferative activity of thecal compartment cells.


Assuntos
Fator de Crescimento Neural/farmacologia , Folículo Ovariano/citologia , Células Tecais/efeitos dos fármacos , Androstenodiona/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Gonadotropina Coriônica/farmacologia , Ciclo-Oxigenase 2 , Dinoprostona/metabolismo , Feminino , Expressão Gênica , Imuno-Histoquímica , Hibridização In Situ , Isoenzimas/genética , Fator de Crescimento Neural/genética , Ovulação/efeitos dos fármacos , Progesterona/metabolismo , Prostaglandina-Endoperóxido Sintases/genética , Sondas RNA , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptor de Fator de Crescimento Neural/genética , Receptor trkA/genética , Receptor trkA/fisiologia , Receptores de Fator de Crescimento Neural/genética , Ovinos , Células Tecais/citologia , Células Tecais/fisiologia
8.
Mol Cell Endocrinol ; 163(1-2): 67-71, 2000 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-10963876

RESUMO

Neurotrophins (NTs) and their receptors play an essential role in the differentiation and survival of defined neuronal populations of the central and peripheral nervous systems. Their actions, however, do not appear to be limited to the nervous system, as both NTs and their receptors have been found in non neuronal cells, including cells of the endocrine system. At least four of the five known neurotrophins, including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4), and their receptors (p75 NTR, trkA, trkB and trkC) are present in the developing ovary. Using mice carrying null mutations of the genes encoding neurotrophins (NGF, NT-4, BDNF) or the receptor that mediates the actions of NT-4 and BDNF (trkB), we have obtained initial results consistent with the notion that neurotrophins are required for the growth of primordial follicles. NGF-deficient mice show a decreased formation of both primary and secondary preantral follicles. Null mutation of the NT-4 gene failed to affect either folliculogenesis or follicular development. However, formation of primary and secondary follicles was compromised in mice carrying a null mutation of both the NT-4 and BDNF genes, suggesting compensation of function by BDNF in NT-4 knockouts. Support for this concept is provided by the similar deficiency in follicular growth observed in animals carrying a null mutation of the gene encoding trkB, the receptors mediating NT-4 and BDNF actions. Initial experiments, using differential display, to isolate genes that may be involved in the process of folliculogenesis and/or early follicular development, resulted in the isolation of a recently identified cell adhesion molecule and a novel transcription factor originally shown to induce cell transformation. It thus appears that formation and development of mammalian follicles requires the concerted action of genes originally thought to be only involved in cell differentiation/survival of neuronal cells, and genes that may control the growth, differentiation, and cell-cell interactions of somatic and germ cells in the ovary.


Assuntos
Fatores de Crescimento Neural/fisiologia , Folículo Ovariano/fisiologia , Receptores de Fator de Crescimento Neural/fisiologia , Transdução de Sinais , Animais , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Feminino , Fator de Crescimento Neural/fisiologia , Fatores de Crescimento Neural/genética , Neurotrofina 3/fisiologia , Folículo Ovariano/inervação , Receptor de Fator de Crescimento Neural/fisiologia , Receptor trkA/fisiologia , Receptor trkB/fisiologia
9.
Endocrinology ; 141(4): 1325-31, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10746635

RESUMO

Alcohol (ALC) use and abuse by adolescents has been rising at an alarming rate. Whether ALC consumption during prepubertal years affects specific hormones and the process of sexual maturation is not known. We used immature female rhesus macaques to assess the effects of ALC on circulating levels of hormones known to be critical for the pubertal process. Ten monkeys averaging 20.3 +/- 0.3 months of age were bled by saphenous vein puncture at 0830 and 2030 h each day for 5 consecutive days to determine baseline levels of GH, insulin-like growth factor I, FSH, LH, estradiol (E2), and leptin. For the next 12 months, each day at 1330 h five monkeys were administered ALC (2 g/kg), and five monkeys were administered an isocaloric sucrose solution via a nasogastric approach. Blood was again collected twice daily on 5 consecutive days at 24, 28, and 32 months for hormone analysis. Food consumption and weight gain were similar for ALC-treated and control animals. The expected night-related increase in serum GH occurred during late juvenile development (28-32 months of age) in control animals, but was suppressed (P < 0.05) in ALC-treated animals. This action was paralleled by a decrease (P < 0.01) in serum insulin-like growth factor I. Serum LH and E2 were also depressed by ALC, with their effects most pronounced at 32 months (LH, P < 0.01; E2, P < 0.001). Serum FSH and leptin were not altered. Although ALC did not affect age at menarche, the interval between subsequent menstruations was lengthened (P < 0.05), thereby showing that ALC affected the development of a regular monthly pattern of menstruation. These results demonstrate the detrimental effects of ALC on the activation of hormone secretion that accompanies puberty in female rhesus monkeys. They also suggest that the subsequent growth spurt and normal timing or progression of puberty may be at risk in human adolescents and teenagers consuming even relatively moderate amounts of ALC on a regular basis.


Assuntos
Consumo de Bebidas Alcoólicas/sangue , Hormônios/metabolismo , Animais , Relação Dose-Resposta a Droga , Etanol/farmacologia , Feminino , Hormônio do Crescimento/sangue , Hormônios/sangue , Macaca mulatta/crescimento & desenvolvimento , Ciclo Menstrual/efeitos dos fármacos , Ciclo Menstrual/fisiologia
10.
Endocrinology ; 141(3): 1059-72, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10698182

RESUMO

A form of polycystic ovary (PCO) resembling some aspects of the human PCO syndrome can be induced in rats by a single injection of estradiol valerate (EV). An increase in sympathetic outflow to the ovary precedes, by several weeks, the appearance of cysts, suggesting the involvement of a neurogenic component in the pathology of this ovarian dysfunction. The present study was carried out to test the hypotheses that this change in sympathetic tone is related to an augmented production of ovarian nerve growth factor (NGF), and that this abnormally elevated production of NGF contributes to the formation of ovarian cysts induced by EV. Injection of the steroid resulted in increased intraovarian synthesis of NGF and its low affinity receptor, p75 NGFR. The increase was maximal 30 days after EV, coinciding with the elevation in sympathetic tone to the ovary and preceding the appearance of follicular cysts. Intraovarian injections of the retrograde tracer fluorogold combined with in situ hybridization to detect tyrosine hydroxylase (TH) messenger RNA-containing neurons in the celiac ganglion revealed that these changes in NGF/p75 NGFR synthesis are accompanied by selective activation of noradrenergic neurons projecting to the ovary. The levels of RBT2 messenger RNA, which encodes a beta-tubulin presumably involved in slow axonal transport, were markedly elevated, indicating that EV-induced formation of ovarian cysts is preceded by functional activation ofceliac ganglion neurons, including those innervating the ovary. Intraovarian administration of a neutralizing antiserum to NGF in conjunction with an antisense oligodeoxynucleotide to p75 NGFR, via Alzet osmotic minipumps, restored estrous cyclicity and ovulatory capacity in a majority of EV-treated rats. These functional changes were accompanied by restoration of the number of antral follicles per ovary that had been depleted by EV and a significant reduction in the number of both precystic follicles and follicular cysts. The results indicate that the hyperactivation of ovarian sympathetic nerves seen in EV-induced PCO is related to an overproduction of NGF and its low affinity receptor in the gland. They also suggest that activation of this neurotrophic-neurogenic regulatory loop is a component of the pathological process by which EV induces cyst formation and anovulation in rodents. The possibility exists that a similar alteration in neurotrophic input to the ovary contributes to the etiology and/or maintenance of the PCO syndrome in humans.


Assuntos
Estradiol/análogos & derivados , Estrogênios Conjugados (USP)/toxicidade , Fatores de Crescimento Neural/biossíntese , Ovário/metabolismo , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/metabolismo , Receptores de Fator de Crescimento Neural/biossíntese , Animais , Anticorpos Bloqueadores/farmacologia , Reagentes de Ligações Cruzadas , Estradiol/toxicidade , Feminino , Imuno-Histoquímica , Hibridização In Situ , Fatores de Crescimento Neural/antagonistas & inibidores , Fatores de Crescimento Neural/imunologia , Norepinefrina/fisiologia , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Ovário/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Fator de Crescimento Neural/antagonistas & inibidores , Ribonucleases/metabolismo , Sistema Nervoso Simpático/fisiologia , Tirosina 3-Mono-Oxigenase/metabolismo
11.
Endocrinology ; 141(3): 1073-82, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10698183

RESUMO

A single injection of estradiol valerate induces a form of cystic ovary resembling some aspects of the human polycystic ovarian syndrome. Preceding the development of follicular cysts, there is an increase in intraovarian synthesis of nerve growth factor (NGF) and the low affinity NGF receptor (p75 NGFR). Selective blockade of NGF actions and p75 NGFR synthesis in the ovary restored estrous cyclicity and ovulatory capacity in estradiol valerate-treated rats, suggesting that an increase in NGF-dependent, p75 NGFR-mediated actions within the ovary contributes to the development of cystic ovarian disease. We have tested this hypothesis by grafting NGF-producing neural progenitor cells into the ovary of juvenile rats that have been induced to ovulate precociously by a single injection of PMSG. The NGF-producing cells, detected by their content of immunoreactive p75 NGFR material, were found scattered throughout the ovary with some of them infiltrating the granulosa cell compartment of large, precystic follicles. Ovarian NGF content was 2-fold higher than in the ovary of rats receiving control cells. Estrous cyclicity was disrupted, with the animals showing prolonged periods of persistent estrus, and an almost continuous background of vaginal cornified cells at other phases of the estrous cycle. Morphometric analysis revealed that the presence of NGF-producing cells neither reduced the total number of corpora lutea per ovary nor significantly increased the formation of follicular cysts. However, the ovaries receiving these cells showed an increased incidence of precystic, type III follicles, accompanied by a reduced number of healthy antral follicles, and an increased size of both healthy and atretic follicles. These changes in follicular dynamics were accompanied by a selective increase in serum androstenedione levels. The results show that an abnormally elevated production of NGF within the ovary suffices to initiate several of the structural and functional alterations associated with the development of follicular cysts in the rat ovary.


Assuntos
Androgênios/metabolismo , Estro/efeitos dos fármacos , Fatores de Crescimento Neural/fisiologia , Ovário/fisiologia , Androgênios/sangue , Androstenodiona/metabolismo , Animais , Transplante de Células/fisiologia , Ensaio de Imunoadsorção Enzimática , Feminino , Técnicas de Transferência de Genes , Imuno-Histoquímica , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia , Ovário/efeitos dos fármacos , Ovário/metabolismo , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/metabolismo , Radioimunoensaio , Ratos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Testosterona/metabolismo , Fatores de Tempo
12.
Mol Hum Reprod ; 6(1): 11-8, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10611255

RESUMO

Although progesterone plays an essential role in ovulation and the luteiniziation of the primate follicle, the expression of cellular components required for progesterone synthesis and their control is not well defined. This study was designed to determine the time course and gonadotrophin versus steroid regulation of the transcription of genes involved in progesterone synthesis in peri-ovulatory follicles. Granulosa cells or whole ovaries were obtained from macaques undergoing controlled ovarian stimulation either before (0 h) or up to 36 h following the administration of an ovulatory human chorionic gonadotrophin (HCG) bolus with or without a 3beta-hydroxysteroid dehydrogenase (3beta-HSD) inhibitor, with or without a non-metabolizable progestin. Granulosa cell concentrations of low density lipoprotein receptor (LDL-R) and steroidogenic acute regulatory protein (StAR) mRNA increased transiently 12 h following HCG administration (P < 0.05) at which time steroid depletion tended to reduce StAR mRNA (P = 0.06). At 36 h post-HCG progesterone suppressed the LDL-R mRNA levels (P < 0.05). P450 side-chain cleavage (P450scc) mRNA decreased in a time-dependent fashion up to 24 h, whereas 3beta-HSD mRNA increased within 12 h of HCG administration (P < 0.05) in a steroid-independent manner. Whole ovarian 17alpha-hydroxylase (P450c17) and granulosa cell P450 aromatase (P450arom) mRNA declined in a time-dependent fashion; by 36 h after HCG administration, steroid depletion increased P450arom mRNA, although progestin replacement did not return aromatase to control values (P < 0.05). These data demonstrate diverse patterns of steroidogenic enzyme expression that generally reflect the conversion of the macaque peri-ovulatory follicle from an oestrogen to progesterone producing gland. Although mRNAs associated with progesterone synthesis and metabolism are primarily regulated by gonadotrophins, cholesterol uptake and utilization may be modulated locally by steroids in luteinizing granulosa cells.


Assuntos
3-Hidroxiesteroide Desidrogenases/genética , Aromatase/genética , Gonadotropina Coriônica/metabolismo , Regulação Enzimológica da Expressão Gênica , Células da Granulosa/enzimologia , Esteroide 17-alfa-Hidroxilase/genética , Animais , Gonadotropina Coriônica/farmacologia , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Macaca mulatta , Ovulação/fisiologia , Fosfoproteínas/genética , Receptores de LDL/genética
13.
Endocrinology ; 138(8): 3320-9, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9231784

RESUMO

The initiation of follicular growth in the mammalian ovary is a gonadotropin-independent phenomenon. Although some of the intraovarian signaling molecules that control the later phases of this process have been recently identified, the factors involved in the acquisition of gonadotropin receptors by early growing follicles have not been fully defined. In the rat, development of the ovarian innervation precedes the onset of folliculogenesis and occurs before follicles acquire responsiveness to gonadotropins. Because vasoactive intestinal polypeptide (VIP) and norepinephrine (NE), two of the neurotransmitters contained in ovarian nerves, are present in the ovary before the gland becomes responsive to gonadotropins, we sought to determine if VIP and/or NE are able to act on early follicles to facilitate the process of molecular differentiation that leads to gonadotropin dependency. In vitro exposure of 2-day-old rat ovaries to isoproterenol (ISO), a beta-adrenoreceptor agonist, or VIP, a neurotransmitter contained in both sympathetic and sensory nerves, increased the steady state levels of the messenger RNAs encoding cytochrome P-450 aromatase (P-450arom) and FSH receptors (FSHR) within 8 h of treatment. A similar effect was observed following forskolin-induced activation of cAMP formation. In situ hybridization experiments revealed that both the P-450arom and FSHR hybridization signals were localized to follicles. The increase in FSHR messenger RNA was accompanied by formation of functional receptor molecules, as demonstrated by the ability of FSH to stimulate cAMP formation in ovaries preexposed to either ISO or VIP, but not in untreated ovaries. The stimulatory effect of ISO and VIP on the formation of FSHR coupled to the cAMP generating system was not reproduced by phenylephrine, an alpha-adrenergic agonist, or secretin, a member of the VIP family not recognized by ovarian VIP receptors. Treatment of VIP-primed ovaries with FSH resulted in follicular growth, demonstrating that exposure of the gland to the neurotransmitter led to the formation of a functional complement of FSH receptors. These results suggest that ovarian nerves, acting via neurotransmitters coupled to the cAMP generating system, contribute to the differentiation process by which newly formed primary follicles acquire FSH receptors and responsiveness to FSH. Follicles that begin to grow in more densely innervated ovarian regions, may have a selective advantage over those not exposed to neurotransmitter-activated, cAMP-dependent signals and, thus, may become more rapidly subjected to gonadotropin control.


Assuntos
Neurotransmissores/fisiologia , Folículo Ovariano/fisiologia , Receptores do FSH/genética , Receptores do FSH/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Animais Recém-Nascidos , Aromatase/análise , Aromatase/genética , Aromatase/metabolismo , Sequência de Bases , Colforsina/farmacologia , Primers do DNA/análise , Primers do DNA/química , Primers do DNA/genética , Feminino , Hormônio Foliculoestimulante/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Hibridização In Situ , Isoproterenol/farmacologia , Norepinefrina/farmacologia , Técnicas de Cultura de Órgãos , Folículo Ovariano/química , Folículo Ovariano/metabolismo , Reação em Cadeia da Polimerase , Gravidez , RNA Mensageiro/análise , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores do FSH/análise , Peptídeo Intestinal Vasoativo/farmacologia
14.
Endocrinology ; 137(12): 5662-70, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8940397

RESUMO

Activation of trkA, the nerve growth factor (NGF) tyrosine kinase receptor, has been recently implicated in the process of mammalian ovulation. During the hour preceding follicular rupture, a marked increase in trkA and NGF gene expression occurs in thecal-interstitial cells of the ovary. Immunoneutralization of NGF actions or pharmacological blockade of trkA transducing activity inhibits ovulation, suggesting that activation of the NGF-trkA complex in nonneural cells of the periovulatory follicle is a physiological component of the ovulatory cascade. As thecal cells of Graafian follicles are functionally coupled by gap junctions, and the ovulatory rupture requires dissociation of thecal cell-cell communication, we sought to determine whether NGF affects the integrity of this communication. We now report that NGF-induced activation of trkA receptors in isolated ovarian thecal cells disrupts cell to cell communication by affecting the functional integrity of gap junctions. Bovine thecal cells expressing trkA receptors, but not cells lacking the receptors, respond to NGF with a reduction in the transfer of calcein, a fluorescent dye that passes through gap junctions. This effect was associated with a rapid (10-30 min) increase in serine phosphorylation of connexin-43, the main protein constituent of gap junctions in the ovary. The reduction in dye transfer was not observed when the cells were exposed to epidermal growth factor or other neurotrophins, including neurotrophin 3, neurotrophin 4, and brain-derived neurotrophic factor. Thus, cell-specific activation of trkA receptors in periovulatory follicles may provide one of the signals involved in inducing the cellular dissociation of the follicular wall that precedes ovulatory rupture.


Assuntos
Comunicação Celular , Junções Comunicantes/fisiologia , Fatores de Crescimento Neural/fisiologia , Ovulação/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fator de Crescimento Neural/fisiologia , Células Tecais/fisiologia , Animais , Bovinos , Comunicação Celular/efeitos dos fármacos , Conexina 43/química , Conexina 43/metabolismo , Feminino , Fatores de Crescimento Neural/farmacologia , Folículo Ovariano/efeitos dos fármacos , Ovário/citologia , Ovário/metabolismo , Fosforilação , Receptor trkA/metabolismo
16.
Endocrinology ; 137(1): 198-209, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8536613

RESUMO

Several members of the neurotrophin (NT) family, including nerve growth factor (NGF), NT-3, and NT-4/5, are expressed in the mammalian ovary. As their respective receptor tyrosine kinases are also found in the gland, the possibility exists that NTs act directly on the gonads to exert effects unrelated to their support of the ovarian innervation. We now report that trkA, the NGF receptor tyrosine kinase, is involved in the acute activational process that leads to the first ovulation. The trkA gene becomes transiently expressed in periovulatory follicules at the time of the first preovulatory surge of gonadotropins at puberty; the increase in trkA messenger RNA (mRNA) content is dramatic ( > 100-fold), but transient (approximately 9 h). No such changes in trkB or trkC mRNA were observed; the abundance of these mRNAs, which encode the receptor tyrosine kinase for NT-4/5 and brain-derived neurotrophic factor, and NT-3, respectively, remained at very low levels throughout puberty. In vivo and in vitro experiments demonstrated that the activation of trkA gene expression is brought about by the proestrous discharge of LH. The increase in trkA mRNA levels is mainly localised to cells of the follicular wall and interstitial tissue of the ovary. NGF mRNA abundance also increases at proestrus, with peak values detected about 5 h before ovulation; as in the case of trkA mRNA, NGF mRNA was found in thecal-interstitial cells. Both trkA and NGF protein, detected by immunohistochemistry, were localized to this same ovarian compartment. Interleukin-1 beta (IL-1 beta), a putative mediator of LH action, enhances both trkA and NGF gene expression in ovarian cells, an effect prevented by IL-1ra, a natural IL-1 beta receptor antagonist. Il-1 beta also stimulates PGE2 release, and this effect was inhibited by both NGF antibodies and a trk receptor blocker, NGF antibodies administered in vivo attenuated the increase in ovarian PGE2 synthesis that antedates ovulation. Immunoneutralization of NGF action or pharmacological blockade of trk tyrosine kinase activity targeted to one ovary resulted in the ipsilateral inhibition of ovulation. The remarkably narrow time frame of trkA gene activation at the completion of follicular growth suggests that NGF acting as a neuroendocrinotrophic factor in a developmentally restricted manner contributes to the acute cytodifferentiation process that leads to the first ovulation in mammals.


Assuntos
Ovulação/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fator de Crescimento Neural/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo , Células Cultivadas , Feminino , Gonadotropinas Equinas/farmacologia , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/genética , Neurotensina/genética , Ovário/citologia , Ovário/metabolismo , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/genética , Receptor do Fator Neutrófico Ciliar , Receptor trkA , Receptor trkC , Receptores de Fator de Crescimento Neural/genética
17.
Endocrinology ; 136(10): 4681-92, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7664689

RESUMO

An emerging body of evidence suggests that neurotrophins not only promote neuronal survival and differentiation, but can also target nonneuronal cells for their actions. Neurotrophins initiate their biological effects by binding to cell membrane tyrosine kinase receptors of the trk protooncogene family. In addition, all neurotrophins recognize with similar affinity a different receptor molecule known as p75 nerve growth factor receptor (p75 NGFR) or low affinity NGFR, which appears to interact with the trk receptors to potentiate their response to neurotrophins. The mature mammalian ovary has been shown to synthesize several neurotrophins, including nerve growth factor (NGF), neurotrophin 3 (NT-3), and neurotrophin 4/5 (NT-4/5). The ovary also expresses some of the neurotrophin receptors, including p75 NGFR, trkB [the receptor for NT-4/5 and brain-derived neurotropic factor (BDNF)], and trkA (the NGF receptor). The present experiments were undertaken to determine whether neurotrophins and their receptors are expressed at the time of definitive ovarian histogenesis, and whether any of them exhibit a developmental pattern of expression related to the completion of folliculogenesis. Immunohistochemical identification of p75 NGFR in rat embryonic ovaries revealed that the receptor is predominantly expressed in mesenchymal cells. By gestational day 18, these cells have formed pockets that enclose presumptive pregranulosa cells and groups of oocytes into ovigerous cords. Immediately after birth, the ovigerous cords are subdivided, resulting in the abrupt formation of primordial follicles between 24-48 h after birth. Consistent with these observations, the p75 NGFR messenger RNA (mRNA) content increased after birth and remained elevated at the time of follicular assembly. The NGF and trkA genes showed a different pattern of expression, as the ovarian content of both NGF and trkA mRNA decreased at the time of folliculogenesis. In contrast to the drop in NGF and trkA mRNA expression, NT-4 mRNA levels increased at the time of follicular assembly, coinciding with the abrupt appearance of trkB mRNA. In situ hybridization showed that the increase in NT-4 mRNA expression occurred in a subpopulation of oocytes between 24-48 h after birth, and that the trkB gene became predominantly expressed at this time in epithelial pregranulosa cells. Substantial, but unchanging, levels of NT-3 mRNA and the mRNA encoding trkC, the preferred NT-3 receptor, were detected throughout the perinatal period examined. Very low and invariable levels of BDNF were also detected.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fatores de Crescimento Neural/genética , Folículo Ovariano/fisiologia , Ovário/metabolismo , Receptores de Fator de Crescimento Neural/genética , Animais , Feminino , Neurotrofina 3 , Proteínas Proto-Oncogênicas/genética , Ratos , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/genética , Receptor trkA , Receptor trkB , Receptor trkC , Receptores de Fator de Crescimento Neural/análise
18.
J Neurosci ; 15(6): 4223-37, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7790907

RESUMO

Hypothalamic neurons control a variety of important hormonal and behavioral functions. Little is known, however, about the neurotrophic factors that these neurons may require for survival and/or maintenance of their differentiated functions. We conducted experiments to examine this issue, utilizing a combination of immunohistochemical, in situ hybridization and cell culture approaches. We found that the low affinity receptor for nerve growth factor (p75 NGFR) is present in small subsets of hypothalamic peptidergic neurons identified as such by their content of galanin, luteinizing hormone-releasing hormone (LHRH) and vasointestinal peptide (VIP). More prominently, however, examination of hypothalamic dopaminergic (DA) neurons for the presence of p75 NGFR-like immunoreactivity revealed that the receptor was present on tyrosine hydroxylase (TH)-positive neurons of the zona incerta and periventricular region, but not on neuroendocrine DA neurons of the tuberoinfundibular region. In situ hybridization experiments using a p75 NGFR cRNA confirmed this distribution. Regardless of the presence or absence of p75 NGFR, neither DA group expresses trkA mRNA, indicating that these two major hypothalamic subsets of DNA neurons are NGF-insensitive. A substantial fraction of TH mRNA-positive cells in the zona incerta expresses trkB mRNA, which encodes the receptor for brain derived neurotrophic factor (BDNF); in turn BDNF supports the in vitro survival of hypothalamic TH neurons bearing p75-NGFR, suggesting that BDNF is trophic for DNA neurons of the zona incerta. In contrast, tuberoinfundibular DA neurons do not express trkB mRNA, but some have trkC mRNA, which encodes the receptor for neurotrophin-3 (NT-3). The in vitro survival of TH neurons devoid of p75-NGFR is supported by NT-3, implying that NT-3 may be trophic for a subset of tuberoinfundibular DA neurons. These results suggest that, in spite of expressing an identical neurotransmitter phenotype, anatomically and functionally segregated DA neurons of the neurodendocrine brain are sustained by different neurotrophic factors.


Assuntos
Hipotálamo/anatomia & histologia , Hipotálamo/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Sistemas Neurossecretores/anatomia & histologia , Sistemas Neurossecretores/fisiologia , Prosencéfalo/fisiologia , Proteínas Proto-Oncogênicas/biossíntese , Receptores Proteína Tirosina Quinases/biossíntese , Receptores de Fator de Crescimento Neural/biossíntese , Animais , Anticorpos Monoclonais , Feminino , Expressão Gênica , Imuno-Histoquímica , Especificidade de Órgãos , Prosencéfalo/anatomia & histologia , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/análise , Receptores Proteína Tirosina Quinases/metabolismo , Receptor trkA , Receptores de Fator de Crescimento Neural/análise , Receptores de Fator de Crescimento Neural/metabolismo
19.
Endocrinology ; 134(3): 1146-54, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8119153

RESUMO

When the ovaries of 23-day-old juvenile rats are transplanted to an ectopic site, they recover within 1 week the ability to control gonadotropin secretion via steroid negative feedback. Vascular corrosion casting followed by scanning electron microscopy revealed that the transplanted ovary becomes profusely revascularized within 48 h after transplantation. Vascular ingrowth was accompanied by a 40- to 60-fold increase in expression of the genes encoding two angiogenic factors, vascular endothelial growth factor (VEGF) and transforming growth factor-beta 1 (TGF beta 1), as assessed by RNA blot hybridization of the corresponding mRNAs. Although TGF beta 3 mRNA levels also increased, no changes in the levels of mRNAs encoding other putative angiogenic factors, such as TGF alpha, basic fibroblast growth factor, and TGF beta 2, were observed. Hybridization histochemistry demonstrated that in intact ovaries, VEGF mRNA is mainly expressed in granulosa cells of the cumulus oophorus and thecal cells of large antral follicles. Transplantation is followed by an increase in mRNA abundance and a dramatic shift in cellular localization, so that the mRNA becomes predominantly expressed in cells of the outer ovarian cortex. In intact ovaries, low levels of TGF beta 1 mRNA were detected in thecal-interstitial cells; after transplantation, its expression also became more predominant in the ovarian outer cortex, but this change was not as marked as in the case of VEGF. Because ovarian autotransplantation is followed by a rapid increase in serum gonadotropin levels, experiments were conducted to determine the importance of this rise in the activation of VEGF and TGF beta 1 gene expression. After transplantation, some animals were treated with the LHRH antagonist Nal-Glu LHRH (50 micrograms/rat, once a day for 2 days) to prevent the posttransplantation rise in serum gonadotropins. Quantitation of VEGF and TGF beta 1 mRNA by RNase protection assay 48 h later showed that suppression of gonadotropin secretion diminished the increase in both VEGF and TGF beta 1 gene expression. Concomitant treatment with PMSG (8 IU/rat, single injection), which mainly bypasses the suppression of endogenous FSH levels, restored the TGF beta 1 mRNA response, but had no effect on VEGF mRNA. The results suggest that the increase in gonadotropin secretion following ovarian transplantation contributes to revascularization of the graft by up-regulating the gene expression of two major angiogenic factors.


Assuntos
Indutores da Angiogênese/genética , Fatores de Crescimento Endotelial/genética , Gonadotropinas/fisiologia , Linfocinas/genética , Ovário/irrigação sanguínea , Ovário/transplante , Fator de Crescimento Transformador beta/genética , Animais , Feminino , Regulação da Expressão Gênica , Gonadotropinas/sangue , Ovário/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Transplante Autólogo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
20.
Front Neuroendocrinol ; 13(2): 120-62, 1992 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1468600

RESUMO

The concept is proposed that polypeptide neurotrophic factors contribute to the developmental regulation of ovarian and hypothalamic function in mammals. Nerve growth factor (NGF) and neurotrophin-3, two members of the neurotrophin family, have been identified in the rat ovary and one of its receptors has been localized to the innervation and thecal cells of developing follicles. Although NGF supports the sympathetic innervation of the gland, the extent to which follicles are innervated appears to be defined by the differential expression of NGF receptors in the theca of developing follicles. The presence of NGF receptors in steroid-producing cells suggests a direct involvement of neurotrophins in the regulation of gonadal endocrine function. Evidence is beginning to emerge suggesting that development of the reproductive hypothalamus is affected by insulin-like growth factor 1 secreted by peripheral tissues, and transforming growth factor alpha (TGF alpha) produced locally. In the rat hypothalamus, TGF alpha appears to be synthesized in both neurons and glial cells. In glial cells it may interact with epidermal growth factor (EGF) receptors to further enhance TGF alpha synthesis and to, perhaps, stimulate eicosanoid formation. In turn, one of these eicosanoids, prostaglandin E2, may act on luteinizing hormone-releasing hormone (LHRH) neurons to stimulate the release of LHRH in a genomic-independent manner. This provides the basis for the notion that during development LHRH secretion is regulated by a dual mechanism, one that involves transsynaptic effects exerted by neurotransmitters, the other that requires a glial-neuronal interaction and that may predominantly regulate release of the neuropeptide. An increased expression of the TGF alpha and EGF receptor genes in reactive astrocytes is postulated to contribute to the process by which hypothalamic injury causes sexual precocity. Morphological maturation of the reproductive hypothalamus is thought to occur during sexual development. The process is accelerated by estradiol, which exerts its neurotrophic effects by enhancing the expression of genes encoding cytoskeletal proteins involved in neuronal development and regeneration. It is suggested that acquisition of functional competence by both the ovaries and the reproductive hypothalamus requires the participation of specific, but not similar, neurotrophic factors. The relevance of these concepts to the process of sexual development in other species, particularly primates, remains to be defined.


Assuntos
Hipotálamo/crescimento & desenvolvimento , Família Multigênica/genética , Fatores de Crescimento Neural/fisiologia , Ovário/crescimento & desenvolvimento , Maturidade Sexual/fisiologia , Animais , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/citologia , Neuroglia/fisiologia , Neurônios/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA