Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 14(4): e0215389, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31026287

RESUMO

Progesterone membrane receptor component 1 (Pgrmc1) is a cytochrome b5-related protein with wide-ranging functions studied most extensively in non-neural tissues. We previously demonstrated that Pgrmc1 is widely distributed in the brain with highest expression in the limbic system. To determine Pgrmc1 functions in cells of these regions, we compared transcriptomes of control siRNA-treated and Pgrmc1 siRNA-treated N42 hypothalamic cells using whole genome microarrays. Our bioinformatics analyses suggested that Pgrmc1 plays a role in immune functions and likely regulates proinflammatory cytokine signaling. In follow-up studies, we showed that one of these cytokines, TNFα, increased expression of rtp4, ifit3 and gbp4, genes found on microarrays to be among the most highly upregulated by Pgrmc1 depletion. Moreover, either Pgrmc1 depletion or treatment with the Pgrmc1 antagonist, AG-205, increased both basal and TNFα-induced expression of these genes in N42 cells. TNFα had no effect on levels of Rtp4, Ifit3 or Gbp4 mRNAs in mHippoE-18 hippocampal control cells, but Pgrmc1 knock-down dramatically increased basal and TNFα-stimulated expression of these genes. P4 had no effect on gbp4, ifit3 or rtp4 expression or on the ability of Pgrmc1 to inhibit TNFα induction of these genes. However, a majority of the top upstream regulators of Pgrmc1 target genes were related to synthesis or activity of steroids, including P4, that exert neuroprotective effects. In addition, one of the identified Pgrmc1 targets was Nr4a1, an orphan receptor important for the synthesis of most steroidogenic molecules. Our findings indicate that Pgrmc1 may exert neuroprotective effects by suppressing TNFα-induced neuroinflammation and by regulating neurosteroid synthesis.


Assuntos
Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Receptores de Progesterona/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Hipotálamo/citologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Camundongos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Progesterona/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores de Progesterona/antagonistas & inibidores , Receptores de Progesterona/genética , Regulação para Cima
2.
PLoS One ; 13(10): e0206570, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30372469

RESUMO

The process of selecting students likely to complete science, technology, engineering and mathematics (STEM) doctoral programs has not changed greatly over the last few decades and still relies heavily on Graduate Record Examination (GRE) scores in most U.S. universities. It has been long debated whether the GRE is an appropriate selection tool and whether overreliance on GRE scores may compromise admission of students historically underrepresented in STEM. Despite many concerns about the test, there are few studies examining the efficacy of the GRE in predicting PhD completion and even fewer examining this question in STEM fields. For the present study, we took advantage of a long-lived collaboration among institutions in the Northeast Alliance for Graduate Education and the Professoriate (NEAGEP) to gather comparable data on GRE scores and PhD completion for 1805 U.S./Permanent Resident STEM doctoral students in four state flagship institutions. We found that GRE Verbal (GRE V) and GRE Quantitative (GRE Q) scores were similar for women who completed STEM PhD degrees and those who left programs. Remarkably, GRE scores were significantly higher for men who left than counterparts who completed STEM PhD degrees. In fact, men in the lower quartiles of GRE V or Q scores finished degrees more often than those in the highest quartile. This pattern held for each of the four institutions in the study and for the cohort of male engineering students across institutions. GRE scores also failed to predict time to degree or to identify students who would leave during the first year of their programs. Our results suggests that GRE scores are not an effective tool for identifying students who will be successful in completing STEM doctoral programs. Considering the high cost of attrition from PhD programs and its impact on future leadership for the U.S. STEM workforce, we suggest that it is time to develop more effective and inclusive admissions strategies.


Assuntos
Teste de Admissão Acadêmica , Educação de Pós-Graduação/normas , Escolaridade , Ciência/educação , Teste de Admissão Acadêmica/estatística & dados numéricos , Educação de Pós-Graduação/estatística & dados numéricos , Engenharia/educação , Engenharia/normas , Feminino , Humanos , Masculino , Matemática/educação , Matemática/normas , Ciência/normas , Fatores Sexuais , Evasão Escolar/estatística & dados numéricos , Tecnologia/educação , Tecnologia/normas , Estados Unidos
3.
Biol Reprod ; 99(2): 433-445, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30101293

RESUMO

The pubertal transition of gonadotropin secretion in pigs is metabolically gated. Kisspeptin (KISS1) and neurokinin B (NKB) are coexpressed in neurons within the arcuate nucleus of the hypothalamus (ARC) and are thought to play an important role in the integration of nutrition and metabolic state with the reproductive neuroendocrine axis. The hypothesis that circulating concentrations of luteinizing hormone (LH) and expression of KISS1 and tachykinin 3(TAC3, encodes NKB) in the ARC of female pigs are reduced with negative energy balance was tested using ovariectomized, prepubertal gilts fed to either gain or lose body weight. Restricted feeding of ovariectomized gilts caused a rapid and sustained metabolic response characterized by reduced concentrations of plasma urea nitrogen, insulin, leptin, and insulin-like growth factor-1 and elevated concentrations of free fatty acids. The secretory pattern of LH shifted from one of low amplitude to one of high amplitude, which caused overall circulating concentrations of LH to be greater in restricted gilts. Nutrient-restricted gilts had greater expression of follicle-stimulating hormone and gonadotropin-releasing hormone receptor, but not LH in the anterior pituitary gland. Expression of KISS1 in the ARC was not affected by dietary treatment, but expression of TAC3 was greater in restricted gilts. These data are consistent with the idea that hypothalamic expression of KISS1 is correlated with the number of LH pulse in pig, and further indicate that amplitude of LH pulses may be regulated by NKB in the gilt.


Assuntos
Metabolismo Energético/fisiologia , Privação de Alimentos/fisiologia , Hipotálamo/metabolismo , Hormônio Luteinizante/metabolismo , Neurocinina B/metabolismo , Adeno-Hipófise/metabolismo , Animais , Ácidos Graxos não Esterificados/sangue , Feminino , Hormônio Foliculoestimulante/metabolismo , Insulina/sangue , Kisspeptinas/metabolismo , Leptina/sangue , Neurônios/metabolismo , Receptores LHRH/metabolismo , Suínos
4.
Neuroscience ; 329: 239-53, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27185484

RESUMO

Developmental exposure to arylhydrocarbon receptor (AhR) ligands abolishes sex differences in a wide range of neural structures and functions. A well-studied example is the anteroventral periventricular nucleus (AVPV), a structure that controls sex-specific luteinizing hormone (LH) release. In the male, testosterone (T) secreted by the developing testes defeminizes LH release mechanisms; conversely, perinatal AhR activation by 2,3,7,8,-tetrachlorodibenzo-p-dioxin (TCDD) blocks defeminization. To better understand developmental mechanisms altered by TCDD exposure, we first verified that neonatal TCDD exposure in male rats prevented the loss of AVPV GABA/glutamate neurons that are critical for female-typical LH surge release. We then used whole genome arrays and quantitative real-time polymerase chain reaction (QPCR) to compare AVPV transcriptomes of males treated neonatally with TCDD or vehicle. Our bioinformatics analyses showed that TCDD enriched gene sets important for neuron development, synaptic transmission, ion homeostasis, and cholesterol biosynthesis. In addition, upstream regulatory analysis suggests that both estrogen receptors (ER) and androgen receptors (AR) regulate genes targeted by TCDD. Of the 23 mRNAs found to be changed by TCDD at least 2-fold (p<0.05), most participate in the functions identified in our bioinformatics analyses. Several, including matrix metallopeptidase 9 and SRY-box 11 (Sox11), are known targets of E2. CUG triplet repeat, RNA binding protein 2 (cugbp2) is particularly interesting because it is sex-specific, oppositely regulated by estradiol (E2) and TCDD. Moreover, it regulates the post-transcriptional processing of molecules previously linked to sexual differentiation of the brain. These findings provide new insights into how TCDD may interfere with defeminization of LH release patterns.


Assuntos
Hipotálamo Anterior/efeitos dos fármacos , Hormônio Luteinizante/metabolismo , Neurônios/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Caracteres Sexuais , Transcriptoma/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Contagem de Células , Ácido Glutâmico/metabolismo , Hipotálamo Anterior/crescimento & desenvolvimento , Hipotálamo Anterior/metabolismo , Masculino , Neurônios/metabolismo , Distribuição Aleatória , Ratos Sprague-Dawley , Ácido gama-Aminobutírico/metabolismo
5.
Front Neurosci ; 7: 164, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-24065878

RESUMO

Progesterone (P4) regulates a wide range of neural functions and likely acts through multiple receptors. Over the past 30 years, most studies investigating neural effects of P4 focused on genomic and non-genomic actions of the classical progestin receptor (PGR). More recently the focus has widened to include two groups of non-classical P4 signaling molecules. Members of the Class II progestin and adipoQ receptor (PAQR) family are called membrane progestin receptors (mPRs) and include: mPRα (PAQR7), mPRß (PAQR8), mPRγ (PAQR5), mPRδ (PAQR6), and mPRε (PAQR9). Members of the b5-like heme/steroid-binding protein family include progesterone receptor membrane component 1 (PGRMC1), PGRMC2, neudesin, and neuferricin. Results of our recent mapping studies show that members of the PGRMC1/S2R family, but not mPRs, are quite abundant in forebrain structures important for neuroendocrine regulation and other non-genomic effects of P4. Herein we describe the structures, neuroanatomical localization, and signaling mechanisms of these molecules. We also discuss possible roles for Pgrmc1/S2R in gonadotropin release, feminine sexual behaviors, fluid balance and neuroprotection, as well as catamenial epilepsy.

6.
Front Neuroendocrinol ; 33(1): 36-44, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21741397

RESUMO

Sex differences in luteinizing hormone (LH) release patterns are controlled by the hypothalamus, established during the perinatal period and required for fertility. Female mammals exhibit a cyclic surge pattern of LH release, while males show a tonic release pattern. In rodents, the LH surge pattern is dictated by the anteroventral periventricular nucleus (AVPV), an estrogen receptor-rich structure that is larger and more cell-dense in females. Sex differences result from mitochondrial cell death triggered in perinatal males by estradiol derived from aromatization of testosterone. Herein we provide an historical perspective and an update describing evidence that molecules important for cell survival and cell death in the immune system also control these processes in the developing AVPV. We conclude with a new model proposing that development of the female AVPV requires constitutive activation of the Tnfα, Tnf receptor 2, NfκB and Bcl2 pathway that is blocked by induction of Tnf receptor-associated factor 2-inhibiting protein (Traip) in the male.


Assuntos
Núcleo Hipotalâmico Anterior/crescimento & desenvolvimento , Núcleos Anteriores do Tálamo/crescimento & desenvolvimento , Hormônio Luteinizante/metabolismo , NF-kappa B/fisiologia , Diferenciação Sexual/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Núcleo Hipotalâmico Anterior/fisiologia , Núcleos Anteriores do Tálamo/fisiologia , Morte Celular , Feminino , Masculino , Mitocôndrias , Fator 2 Associado a Receptor de TNF/antagonistas & inibidores , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/fisiologia
7.
J Comp Neurol ; 519(17): 3456-69, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21800299

RESUMO

It is now well established that the kisspeptin neurons of the hypothalamus play a key role in regulating the activity of gonadotropin-releasing hormone (GnRH) neurons. The population of kisspeptin neurons residing in the rostral periventricular region of the third ventricle (RP3V), encompassing the anteroventral periventricular (AVPV) and periventricular preoptic nuclei (PVpo), are implicated in the generation of the preovulatory GnRH surge mechanism and puberty onset in female rodents. The present study examined whether these kisspeptin neurons may express other neuropeptides in the adult female mouse. Initially, the distribution of galanin, neurotensin, met-enkephalin (mENK), and cholecystokinin (CCK)-immunoreactive cells was determined within the RP3V of colchicine-treated mice. Subsequent experiments, using a new kisspeptin-10 antibody raised in sheep, examined the relationship of these neuropeptides to kisspeptin neurons. No evidence was found for expression of neurotensin or CCK by RP3V kisspeptin neurons, but subpopulations of kisspeptin neurons were observed to express galanin and mENK. Dual-labeled RP3V kisspeptin/galanin cells represented 7% of all kisspeptin and 21% of all galanin neurons whereas dual-labeled kisspeptin/mENK cells represented 28-38% of kisspeptin neurons and 58-68% of the mENK population, depending on location within the AVPV or PVpo. Kisspeptin neurons in the arcuate nucleus were also found to express galanin but not mENK. These observations indicate that, like the kisspeptin population of the arcuate nucleus, kisspeptin neurons in the RP3V also co-express a range of neuropeptides. This pattern of co-expression should greatly increase the dynamic range with which kisspeptin neurons can modulate the activity of their afferent neurons.


Assuntos
Encefalina Metionina/biossíntese , Galanina/biossíntese , Regulação da Expressão Gênica , Hipotálamo/metabolismo , Kisspeptinas/biossíntese , Neurônios/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/química , Núcleo Arqueado do Hipotálamo/metabolismo , Feminino , Hipotálamo/química , Camundongos , Neurônios/química , Terceiro Ventrículo/química , Terceiro Ventrículo/metabolismo
8.
J Neurosci ; 31(7): 2421-30, 2011 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21325509

RESUMO

The anteroventral periventricular nucleus (AVPV) is thought to play a key role in regulating the excitability of gonadotropin-releasing hormone (GnRH) neurons that control fertility. Using an angled, parahorizontal brain slice preparation we have undertaken a series of electrophysiological experiments to examine how the AVPV controls GnRH neurons in adult male and female mice. More than half (59%) of GnRH neurons located in the rostral preoptic area were found to receive monosynaptic inputs from the AVPV in a sex-dependent manner. AVPV stimulation frequencies <1 Hz generated short-latency action potentials in GnRH neurons with GABA and glutamate mediating >90% of the evoked fast synaptic currents. The AVPV GABA input was dominant and found to excite or inhibit GnRH neurons in a cell-dependent manner. Increasing the AVPV stimulation frequency to 5-10 Hz resulted in the appearance of additional poststimulus inhibitory as well as delayed excitatory responses in GnRH neurons that were independent of ionotropic amino acid receptors. The inhibition observed immediately following the end of the stimulation period was mediated partly by GABA(B) receptors, while the delayed activation was mediated by the neuropeptide kisspeptin. The latter response was essentially absent in Gpr54 knock-out mice and abolished by a Gpr54 antagonist. Together, these studies show that AVPV neurons provide direct amino acid and neuropeptidergic inputs to GnRH neurons. Low-frequency activation generates predominant GABA/glutamate release with higher frequency activation recruiting release of kisspeptin. This frequency-dependent release of amino acid and neuropeptide neurotransmitters greatly expands the range of AVPV control of GnRH neuron excitability.


Assuntos
Aminoácidos/metabolismo , Núcleos Anteriores do Tálamo/citologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Biofísica , Estimulação Elétrica/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Ácido Glutâmico/farmacologia , Hormônio Liberador de Gonadotropina/genética , Proteínas de Fluorescência Verde/genética , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Neurotransmissores/metabolismo , Técnicas de Patch-Clamp , Tempo de Reação/fisiologia , Receptores Acoplados a Proteínas G/deficiência , Receptores de Kisspeptina-1 , Estatísticas não Paramétricas , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Valina/análogos & derivados , Valina/farmacologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Ácido gama-Aminobutírico/metabolismo , Ácido gama-Aminobutírico/farmacologia
9.
Endocrinology ; 152(2): 526-35, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21177834

RESUMO

High levels of circulating prolactin are known to cause infertility, but the precise mechanisms by which prolactin influences the neuroendocrine axis are yet to be determined. We used dual-label in situ hybridization to investigate whether prolactin-receptor (PRLR) mRNA is expressed in GnRH neurons. In addition, because γ-aminobutyric acidergic and kisspeptin neurons in the rostral hypothalamus are known to regulate GnRH neurons and, hence, might mediate the actions of prolactin, we investigated whether these neurons coexpress PRLR mRNA. (35)S-labeled RNA probes to detect PRLR mRNA were hybridized together with digoxigenin-labeled probes to detect either GnRH, Gad1/Gad2, or Kiss1 mRNA in the rostral hypothalamus of ovariectomized (OVX), estradiol-treated rats. Additional sets of serial sections were cut through the arcuate nucleus of OVX rats, without estradiol replacement, to examine coexpression of PRLR mRNA in the arcuate population of kisspeptin neurons. PRLR mRNA was highly expressed throughout the rostral preoptic area, particularly in periventricular regions surrounding the third ventricle, and there was a high degree of colocalization of PRLR mRNA in both Gad1/Gad2 and Kiss1 mRNA-containing cells (86 and 85.5%, respectively). In contrast, only a small number of GnRH neurons (<5%) was found to coexpress PRLR mRNA. In the arcuate nucleus of OVX rats, the majority of Kiss1 mRNA-containing cells also coexpressed PRLR mRNA. These data are consistent with the hypothesis that, in addition to a direct action on a small subpopulation of GnRH neurons, prolactin actions on GnRH neurons are predominantly mediated indirectly, through known afferent pathways.


Assuntos
Fertilidade/fisiologia , Hipotálamo/citologia , Hipotálamo/metabolismo , Neurônios/metabolismo , Proteínas/metabolismo , Receptores da Prolactina/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Feminino , Fertilidade/genética , Glutamato Descarboxilase/genética , Hibridização In Situ , Kisspeptinas , Ovariectomia , Proteínas/genética , Ratos , Ratos Sprague-Dawley , Receptores da Prolactina/genética
10.
Mol Cell Endocrinol ; 333(2): 151-9, 2011 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-21187122

RESUMO

Arylhydrocarbon receptor (Ahr) activation by 2,3,7,8-tetrachlordibenzo-p-dioxin (TCDD) interferes with female reproductive functions, but there is little information on the specific targets of TCDD in the hypothalamic-pituitary-gonadal (HPG) axis. In these studies, we found that TCDD upregulated known AhR target genes, cytochrome p450 1a1 (Cyp1a1), Cyp1a2 and Cyp1b1 in the rat pituitary gland. Moreover, 75% of pituitary lactotropes and 45% of gonadotropes contained Ahr mRNA, and most Ahr-containing cells were estrogen receptor 1 (Esr1)-positive. TCDD abrogated estradiol (E(2))-induced prolactin (Prl) expression in vivo and in vitro; conversely, E(2) blocked TCDD upregulation of luteinizing hormone beta (Lhb) and glycoprotein hormone alpha polypeptide (Cga) expression. TCDD had no effect on levels of Ahr mRNA, but upregulated Esr1 mRNA. E(2) independently repressed Ahr and Esr1 expression and blocked TCDD upregulation of Esr1. Thus, complex interactions between Ahr and Esr alter Prl and luteinizing hormone (LH) synthesis by direct actions in lactotropes and gonadotropes. These findings provide important insights into how TCDD disrupts female reproductive functions.


Assuntos
Estradiol/farmacologia , Subunidade alfa de Hormônios Glicoproteicos/genética , Gonadotrofos/metabolismo , Lactotrofos/metabolismo , Hormônio Luteinizante Subunidade beta/genética , Prolactina/genética , Precursores de Proteínas/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP1B1 , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Subunidade alfa de Hormônios Glicoproteicos/metabolismo , Gonadotrofos/citologia , Gonadotrofos/efeitos dos fármacos , Imuno-Histoquímica , Hibridização In Situ , Lactotrofos/citologia , Lactotrofos/efeitos dos fármacos , Hormônio Luteinizante Subunidade beta/metabolismo , Hipófise/citologia , Hipófise/efeitos dos fármacos , Hipófise/enzimologia , Dibenzodioxinas Policloradas/farmacologia , Prolactina/metabolismo , Precursores de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Hidrocarboneto Arílico/genética
11.
Rev Neurosci ; 21(5): 363-80, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21280455

RESUMO

Estradiol (E2) is a potent neuroactive steroid that acts through both nuclear and membrane estrogen receptors (ER) found widely distributed in the brain. Although long known for its role in the neural control of reproduction, more recent work demonstrates that E2 also affects learning and memory, as well as anxiety and depressive symptoms. These findings prompted studies on the neural consequences of long-term E2 deprivation in postmenopausal women. Despite hundreds of studies in animal models and women, the advisability of hormone replacement therapy (HRT) for neuroprotection remains a contentious issue because the effects of estrogen vary among studies. One difficulty in reconciling the conflicting results is the lack of integration across the neuroscience sub-disciplines that contribute to the field. To address this issue, we first review data on E2 regulation of cognition and mood, as well as on factors that may contribute to the disparate findings across studies. GABA and glutamate are proximal regulators of cognition and mood; therefore we next review review data showing that E2 acts through nuclear- and membrane-initiated mechanisms to regulate GABA and glutamate signaling, respectively. We also review evidence that these E2 signaling mechanisms change with age. Finally, we propose a molecular and cellular model of how E2 can have positive, negative, or no effects on neural functions in the aging brain, and we highlight the current gaps in the literature. Addressing these gaps will facilitate development of the mechanism-based strategies needed for designing more effective HRT regimens.


Assuntos
Encéfalo/fisiologia , Núcleo Celular/metabolismo , Estradiol/metabolismo , Ácido Glutâmico/metabolismo , Terapia de Reposição Hormonal , Transdução de Sinais/fisiologia , Ácido gama-Aminobutírico/metabolismo , Afeto/efeitos dos fármacos , Afeto/fisiologia , Animais , Encéfalo/citologia , Cognição/efeitos dos fármacos , Cognição/fisiologia , Modelos Animais de Doenças , Estradiol/farmacologia , Feminino , Humanos , Masculino
12.
Proc Natl Acad Sci U S A ; 106(39): 16692-7, 2009 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-19805359

RESUMO

Sexually dimorphic brain nuclei underlie gender-specific neural functions and susceptibility to disease, but the developmental basis of dimorphisms is poorly understood. In these studies, we focused on the anteroventral periventricular nucleus (AVPV), a nucleus that is larger in females and critical for the female-typical cyclic surge pattern of luteinizing hormone (LH) release. Sex differences in the size and function of the AVPV result from apoptosis that occurs preferentially in the developing male. To identify upstream pathways responsible for sexual differentiation of the AVPV, we used targeted apoptosis microarrays and in vivo and in vitro follow-up studies. We found that the tumor necrosis factor alpha (TNFalpha)-TNF receptor 2 (TNFR2)-NFkappaB cell survival pathway is active in postnatal day 2 (PND2) female AVPV and repressed in male counterparts. Genes encoding key members of this pathway were expressed exclusively in GABAergic neurons. One gene in particular, TNF receptor-associated factor 2 (TRAF2)-inhibiting protein (trip), was higher in males and it inhibited both TNFalpha-dependent NFkappaB activation and bcl-2 gene expression. The male AVPV also had higher levels of bax and bad mRNA, but neither of these genes was regulated by either TNFalpha or TRIP. Finally, the trip gene was not expressed in the sexually dimorphic nucleus of the preoptic area (SDN-POA), a nucleus in which apoptosis is higher in females than males. These findings form the basis of a new model of sexual differentiation of the AVPV that may also apply to the development of other sexually dimorphic nuclei.


Assuntos
Encéfalo/fisiologia , Diferenciação Sexual , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo , Animais , Núcleo Hipotalâmico Anterior/metabolismo , Feminino , Genes bcl-2 , Masculino , Modelos Biológicos , NF-kappa B/genética , NF-kappa B/metabolismo , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Fator 2 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Ácido gama-Aminobutírico/metabolismo
13.
J Neurosci ; 29(27): 8790-7, 2009 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-19587286

RESUMO

Estradiol (E(2)) regulates a wide range of neural functions, many of which require activation of estrogen receptor alpha (ERalpha) and/or ERbeta, ligand-gated transcriptional regulators. Surprisingly, very few neural gene targets of ERs have been identified, and these cannot easily explain the myriad effects of E(2). GABA regulates most of the same neural functions as E(2), and GABAergic neurons throughout the brain contain ER. Therefore, we examined whether E(2) directly regulates expression of glutamic acid decarboxylase 2 (gad2), the enzyme primarily responsible for GABA synthesis for synaptic release. Using dual luciferase assays, we found that E(2), but not other gonadal steroids, stimulated the activity of a 2691 bp rat gad2 promoter reporter construct. Activation required either ERalpha or ERbeta, and ERbeta did not repress ERalpha-mediated transactivation. Site-directed mutagenesis studies identified three estrogen response elements (EREs) with cell-specific functions. An ERE at -711 upstream of the gad2 translational start site was essential for transactivation in both MCF-7 breast cancer cells and SN56.B5.G4 neural cells, but an ERE at -546 enhanced transcription only in neural cells. A third ERE at -1958 was inactive in neural cells but exerted potent transcriptional repression in E(2)-treated MCF-7 cells. Chromatin immunoprecipitation assays in mouse GABAergic N42 cells confirmed that E(2) induced ERalpha binding to a DNA fragment containing sequences corresponding to the -546 and -711 EREs of the rat promoter. Based on these data, we propose that direct transcriptional regulation of gad2 may explain, at least in part, the ability of E(2) to impact such a diverse array of neural functions.


Assuntos
Estradiol/farmacologia , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Marcação de Genes , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Transcrição Gênica/genética , Animais , Sequência de Bases , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Cães , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Marcação de Genes/métodos , Cavalos , Humanos , Macaca mulatta , Masculino , Camundongos , Dados de Sequência Molecular , Pan troglodytes , Ratos , Salmão
14.
Endocrinology ; 147(6 Suppl): S33-42, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16690800

RESUMO

Historically, much of the research on health effects of environmental pollutants focused on ascertaining whether compounds were carcinogenic. More recent findings show that environmental contaminants also exert insidious effects by disrupting hormone action. Of particular concern are findings that developmental exposure to dioxins, chemicals that act through the aryl hydrocarbon receptor pathway, permanently alters sexually differentiated neural functions in animal models. In this review, we focus on mechanisms through which dioxins disrupt neuroendocrine development as exemplified by effects on a brain region critical for ovulation in rodents. We also provide evidence that dysregulation of GABAergic neural development may be a general mechanism underlying a broad spectrum of effects seen after perinatal dioxin exposure.


Assuntos
Dioxinas/toxicidade , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/embriologia , Receptores de Hidrocarboneto Arílico/fisiologia , Diferenciação Sexual , Animais , Exposição Ambiental , Estradiol/fisiologia , Feminino , Ácido Glutâmico/fisiologia , Humanos , Ligantes , Neurônios/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Reprodução/efeitos dos fármacos , Ácido gama-Aminobutírico/fisiologia
15.
Endocrinology ; 146(10): 4491-9, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16020480

RESUMO

Cannabinoids (CBs) exert untoward effects on reproduction by reducing LH secretion and suppressing gonadal function. Recent evidence suggests these effects are due primarily to hypothalamic dysfunction; however, the mechanism is obscure. Using immortalized hypothalamic GnRH neurons, we find these cells produce and secrete at least two different endocannabinoids. After release, 2-arachidonyl monoacylglycerol and anandamide are rapidly transported into GnRH neurons and are degraded to other lipids by fatty-acid amide hydrolase. The immortalized GnRH neurons also possess CB1 and CB2 receptors that are coupled to Gi/Go proteins whose activation leads to inhibition of GnRH secretion. In perifusion experiments, CBs block pulsatile release of GnRH. When a CB receptor agonist is delivered into the third ventricle of adult female mice, estrous cycles are prolonged by at least 2 d. Although in situ hybridization experiments suggest either that GnRH neurons in vivo do not possess CB1 receptors or that they are very low, transcripts are localized in close proximity to these neurons. Inasmuch as GnRH neurons in vivo possess G protein receptors that are coupled to phospholipase C and increased intracellular Ca2+, these same neurons should also be able to synthesize endocannabinoids. These lipids, in turn, could bind to CB receptors on neighboring cells, and perhaps GnRH neurons, to exert feedback control over GnRH function. This network could serve as a novel mechanism for regulating GnRH secretion where reproductive functions as diverse as the onset of puberty, timing of ovulation, duration of lactational infertility, and initiation/persistence of menopause may be affected.


Assuntos
Canabinoides/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/fisiologia , Receptor CB1 de Canabinoide/fisiologia , Receptor CB2 de Canabinoide/fisiologia , Animais , Linhagem Celular , Hormônio Liberador de Gonadotropina/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/genética , Hipotálamo , Cinética , Macrófagos , Camundongos , Neurônios/efeitos dos fármacos , Receptor CB1 de Canabinoide/efeitos dos fármacos , Receptor CB2 de Canabinoide/efeitos dos fármacos
16.
J Neurosci ; 24(37): 8097-105, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15371511

RESUMO

It is generally assumed that the inhibitory neurotransmitter GABA and the stimulatory neurotransmitter glutamate are released from different neurons in adults. However, this tenet has made it difficult to explain how the same afferent signals can cause opposite changes in GABA and glutamate release. Such reciprocal release is a central mechanism in the neural control of many physiological processes including activation of gonadotropin-releasing hormone (GnRH) neurons, the neural signal for ovulation. Activation of GnRH neurons requires simultaneous suppression of GABA and stimulation of glutamate release, each of which occurs in response to a daily photoperiodic signal, but only in the presence of estradiol (E2). In rodents, E2 and photoperiodic signals converge in the anteroventral periventricular nucleus (AVPV), but it is unclear how these signals differentially regulate GABA and glutamate secretion. We now report that nearly all neurons in the AVPV of female rats express both vesicular glutamate transporter 2 (VGLUT2), a marker of hypothalamic glutamatergic neurons, as well as glutamic acid decarboxylase and vesicular GABA transporter (VGAT), markers of GABAergic neurons. These dual-phenotype neurons are the main targets of E2 in the region and are more than twice as numerous in females as in males. Moreover, dual-phenotype synaptic terminals contact GnRH neurons, and at the time of the surge, VGAT-containing vesicles decrease and VGLUT2-containing vesicles increase in these terminals. Thus, we propose a new model for ovulation that includes dual-phenotype GABA/glutamate neurons as central transducers of hormonal and neural signals to GnRH neurons.


Assuntos
Ácido Glutâmico/análise , Neurônios/classificação , Ovulação/fisiologia , Área Pré-Óptica/citologia , Caracteres Sexuais , Ácido gama-Aminobutírico/análise , Sistemas de Transporte de Aminoácidos/análise , Animais , Biomarcadores , Castração , Ritmo Circadiano/fisiologia , Implantes de Medicamento , Estradiol/farmacologia , Estradiol/fisiologia , Moduladores de Receptor Estrogênico/administração & dosagem , Moduladores de Receptor Estrogênico/farmacologia , Receptor alfa de Estrogênio/biossíntese , Receptor alfa de Estrogênio/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato Descarboxilase/análise , Glutamato Descarboxilase/biossíntese , Glutamato Descarboxilase/genética , Ácido Glutâmico/metabolismo , Hormônio Liberador de Gonadotropina/análise , Hibridização In Situ , Masculino , Proteínas de Membrana Transportadoras/análise , Modelos Biológicos , Terminações Nervosas/química , Terminações Nervosas/ultraestrutura , Proteínas do Tecido Nervoso/análise , Neurônios/química , Neurônios/metabolismo , Neurônios/fisiologia , Fenótipo , Área Pré-Óptica/química , Área Pré-Óptica/fisiologia , Ratos , Ratos Sprague-Dawley , Proteína Vesicular 2 de Transporte de Glutamato , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores , Ácido gama-Aminobutírico/metabolismo
17.
J Comp Neurol ; 473(3): 315-33, 2004 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-15116394

RESUMO

Topographical distribution of estrogen receptor-beta (ER-beta)-synthesizing oxytocin (OT) and vasopressin (VP) neurons was studied in the hypothalamic paraventricular and supraoptic nuclei (PVH; SO) of ovariectomized rats. In distinct subregions, 45-98% of OT neurons and 88-99% of VP neurons exhibited ER-beta immunoreactivity that was confined to cell nuclei. Neuronal populations differed markedly with respect to the intensity of the ER-beta signal. Magnocellular OT neurons in the PVH, SO, and accessory cell groups typically contained low levels of the ER-beta signal; in contrast, robust receptor labeling was displayed by OT cells in the ventral subdivision of medial parvicellular subnucleus and in the caudal PVH (dorsal subdivision of medial parvicellular subnucleus and lateral parvicellular subnucleus). Estrogen receptor-beta signal was generally more intense and present in higher proportions of magnocellular and parvicellular VP vs. OT neurons of similar topography. Immunocytochemical observations were confirmed via triple-label in situ hybridization, an approach combining use of digoxigenin-, fluorescein-, and 35S-labeled cRNA hybridization probes. Further, ER-beta mRNA was also detectable in corticotropin-releasing hormone neurons in the parvicellular PVH. Finally, double-label immunocytochemical analysis of human autopsy samples showed that subsets of OT and VP neurons also express ER-beta in the human. These neuroanatomical studies provide detailed information about the topographical distribution and cellular abundance of ER-beta within subsets of hypothalamic OT and VP neurons in the rat. The variable receptor content may indicate the differential responsiveness to estrogen in distinct OT and VP neuronal populations. In addition, a relevance of these findings to the human hypothalamus is suggested.


Assuntos
Hipotálamo/citologia , Neurônios/metabolismo , Ocitocina/metabolismo , Receptores de Estrogênio/metabolismo , Vasopressinas/metabolismo , Adulto , Idoso , Animais , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Receptor beta de Estrogênio , Feminino , Humanos , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Masculino , Microscopia Eletrônica/métodos , Neurônios/ultraestrutura , Núcleo Hipotalâmico Paraventricular/ultraestrutura , Mudanças Depois da Morte , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos
18.
Biol Reprod ; 69(6): 1771-8, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12890720

RESUMO

Estrogen signaling to GnRH neurons is critical for coordinating the preovulatory surge release of LH with follicular maturation. Until recently it was thought that estrogen signaled GnRH neurons only indirectly through numerous afferent systems. This minireview presents new evidence indicating that GnRH neurons are directly regulated by estradiol (E2), primarily through estrogen receptor (ER)-beta, and indirectly through E2-sensitive neurons in the anteroventral periventricular (AVPV) region. The data described suggest that E2 generally represses GnRH gene expression but that this repression is transiently overcome by indirect E2-dependent signals relayed by AVPV neurons. We also present evidence that the AVPV neurons responsible for relaying E2 signals to GnRH neurons are multifunctional gamma aminobutyric acid-ergic/glutamatergic/neuropeptidergic neurons.


Assuntos
Estradiol/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Animais , Receptor beta de Estrogênio , Feminino , Regulação da Expressão Gênica , Humanos , Receptores de Estrogênio/metabolismo , Transcrição Gênica
19.
Endocrinology ; 143(12): 4837-45, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12446611

RESUMO

Although estradiol (E2) triggers phasic increases in LH-releasing hormone (LHRH) synthesis and release, the neurocircuitry responsible for these changes is unclear. We used an ovariectomized, E2-treated animal model to investigate the possibility that glutamate, through N-methyl-D-aspartate (NMDA) receptors (NMDAR), communicates E2 signals to LHRH neurons. A neuroanatomical analysis of the region containing LHRH neurons revealed that approximately 80% of LHRH neurons in medial, but less than 40% in lateral, nuclei of the preoptic area contained NMDAR1 mRNA. Consistent with this distribution pattern, NMDA doubled LHRH mRNA levels in medial neurons, but increased them by less than 30% in cells of the lateral nuclei. Steroids did not alter NMDAR1 mRNA levels in LHRH neurons or change the percentage of LHRH neurons expressing the gene. Furthermore, in contrast to the regionalized effects of NMDA, E2 treatment increased LHRH mRNA levels to the same extent in medial and lateral neurons, and MK801 failed to block E2-induced changes in LHRH gene expression. These results demonstrate that glutamatergic signaling via NMDA receptors is direct and preferentially targets LHRH neurons in medial nuclei of the preoptic area. However, it is unlikely that NMDAR activation mediates E2-dependent increases in LHRH mRNA levels before the LH surge.


Assuntos
Estradiol/farmacologia , Glutamina/metabolismo , Hormônio Liberador de Gonadotropina/genética , Neurônios/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Transdução de Sinais , Animais , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/análise , Hormônio Liberador de Gonadotropina/fisiologia , N-Metilaspartato/farmacologia , Ovariectomia , Área Pré-Óptica/citologia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/genética , Distribuição Tecidual
20.
J Histochem Cytochem ; 50(10): 1389-400, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12364572

RESUMO

The goal of the present studies was to optimize mRNA detection with radioisotopic in situ hybridization histochemistry (ISHH). Test experiments performed on sections of rat brain tissue used computer-assisted image analysis to compare autoradiographic signals resulting when varying concentrations of (35)S-labeled cRNA probes, dextran sulfate (DS), and dithiothreitol (DTT) were used for ISHH. We found that greatly enhanced corrected signal density (total density of signal area minus background density) was obtained using concentrations of probe and/or DS that were several-fold higher than those widely recommended in published ISHH procedures (probe concentration >4 x 10(4) cpm/microl; DS concentration >10%). Extended hybridization reaction (>16 hr) also significantly augmented the corrected signal density. Finally, nonspecific probe binding was greatly reduced and corrected signal density enhanced by including 750-1000 mM, rather than the widely used 10-200 mM DTT, in the hybridization buffer. These observations indicate that the low efficiency of hybridization and the formation of high background may largely compromise the sensitivity of routine ISHH procedures. We suggest that the new method using increased concentrations of (35)S-labeled cRNA probe, DS, and DTT will be especially important for the cellular localization of rare mRNA species.


Assuntos
Sulfato de Dextrana , Ditiotreitol , Hibridização In Situ/métodos , Sondas RNA , RNA Complementar , Animais , Autorradiografia , Encéfalo/metabolismo , Soluções Tampão , Feminino , Galanina/genética , Galanina/metabolismo , Processamento de Imagem Assistida por Computador , Indicadores e Reagentes , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Radioisótopos de Enxofre
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...