Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Behav Brain Funct ; 11(1): 34, 2015 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-26537115

RESUMO

BACKGROUND: Fibroblast growth factors (FGFs) are crucial signaling molecules that direct the development of the vertebrate brain. FGF8 gene signaling in particular, may be important for the development of the hypothalamus-pituitary-adrenal (HPA)-axis. Indeed, newborn Fgf8 hypomorphic mice harbor a major reduction in the number of vasopressin (VP) neurons in the paraventricular nucleus (PVN), the central output component of the HPA-axis. Additionally, recent studies indicated that adult heterozygous ((+/neo)) Fgf8 hypomorphic mice exhibit more anxiety-like behaviors than wildtype (WT) mice. These studies led us to investigate whether Fgf8 hypomorphy abrogated VP and/or corticotropin-releasing hormone (CRH) neuronal development in the postnatal day (PN) 21 and adult mouse PVN. Furthermore, we studied whether Fgf8 hypomorphy disrupted HPA responsiveness in these mice. METHODS: Using immunohistochemistry, we examined the development of VP and CRH neurons located in the PVN of PN 21 and adult Fgf8 (+/neo) mice. Moreover, we used a restraint stress (RS) paradigm and measured corticosterone levels with enzyme immunoassays in order to assess HPA axis activation. RESULTS: The number of VP neurons in the PVN did not differ between WT and Fgf8 (+/neo) mice on PN 21 and in adulthood. In contrast, CRH immunoreactivity was much higher in Fgf8 (+/neo) mice than in WT mice on PN 21, this difference was no longer shown in adult mice. RS caused a higher increase in corticosterone levels in adult Fgf8 (+/neo) mice than in WT mice after 15 min, but no difference was seen after 45 min. CONCLUSIONS: First, Fgf8 hypomorphy did not eliminate VP and CRH neurons in the mouse PVN, but rather disrupted the postnatal timing of neuropeptide expression onset in PVN neurons. Second, Fgf8 hypomorphy may, in part, be an explanation for affective disorders involving hyperactivity of the HPA axis, such as anxiety.


Assuntos
Fator 8 de Crescimento de Fibroblasto/fisiologia , Células Neuroendócrinas/fisiologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/crescimento & desenvolvimento , Animais , Contagem de Células , Corticosterona/sangue , Hormônio Liberador da Corticotropina/metabolismo , Fator 8 de Crescimento de Fibroblasto/genética , Sistema Hipotálamo-Hipofisário/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Células Neuroendócrinas/citologia , Sistema Hipófise-Suprarrenal/fisiologia , Restrição Física , Vasopressinas/metabolismo
2.
Pflugers Arch ; 465(5): 573-84, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23503727

RESUMO

The concept that the brain differs in make-up between males and females is not new. For example, it is well established that anatomists in the nineteenth century found sex differences in human brain weight. The importance of sex differences in the organization of the brain cannot be overstated as they may directly affect cognitive functions, such as verbal skills and visuospatial tasks in a sex-dependent fashion. Moreover, the incidence of neurological and psychiatric diseases is also highly dependent on sex. These clinical observations reiterate the importance that gender must be taken into account as a relevant possible contributing factor in order to understand the pathogenesis of neurological and psychiatric disorders. Gender-dependent differentiation of the brain has been detected at every level of organization--morphological, neurochemical, and functional--and has been shown to be primarily controlled by sex differences in gonadal steroid hormone levels during perinatal development. In this review, we discuss howthe gonadal steroid hormone testosterone and its metabolites affect downstream signaling cascades, including gonadal steroid receptor activation, and epigenetic events in order to differentiate the brain in a gender-dependent fashion.


Assuntos
Encéfalo/crescimento & desenvolvimento , Epigênese Genética , Caracteres Sexuais , Animais , Encéfalo/metabolismo , Encéfalo/fisiologia , Feminino , Hormônios Esteroides Gonadais/metabolismo , Humanos , Masculino
3.
Am J Physiol Endocrinol Metab ; 303(12): E1428-39, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23047985

RESUMO

The continued presence of gonadotropin-releasing hormone (GnRH) neurons is required for a healthy reproductive lifespan, but factors that maintain postnatal GnRH neurons have not been identified. To begin to understand these factors, we investigated whether 1) fibroblast growth factor (FGF) signaling and 2) interactions with the opposite sex are involved in the maintenance of the postnatal GnRH system. A transgenic mouse model (dnFGFR mouse) with the targeted expression of a dominant-negative FGF receptor (dnFGFR) in GnRH neurons was used to examine the consequence of FGF signaling deficiency on postnatal GnRH neurons. Male dnFGFR mice suffered a significant loss of postnatal GnRH neurons within the first 100 days of life. Interestingly, this loss was reversed after cohabitation with female, but not male, mice for 300-550 days. Along with a rescue in GnRH neuron numbers, opposite-sex housing in dnFGFR males also increased hypothalamic GnRH peptide levels, promoted a more mature GnRH neuronal morphology, facilitated litter production, and enhanced testicular morphology. Last, mice hypomorphic for FGFR3 exhibited a similar pattern of postnatal GnRH neuronal loss as dnFGFR males, suggesting FGF signaling acts, in part, through FGFR3 to enhance the maintenance of the postnatal GnRH system. In summary, we have shown that FGF signaling is required for the continued presence of postnatal GnRH neurons. However, this requirement is not absolute, since sexual interactions can compensate for defects in FGFR signaling, thereby rescuing the declining GnRH system. This suggests the postnatal GnRH system is highly plastic and capable of responding to environmental stimuli throughout adult life.


Assuntos
Envelhecimento , Fator 3 de Crescimento de Fibroblastos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Animais , Contagem de Células , Heterozigoto , Hipotálamo/citologia , Hipotálamo/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Degeneração Neural/etiologia , Degeneração Neural/prevenção & controle , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptores LHRH/metabolismo , Comportamento Sexual Animal , Transmissão Sináptica , Testículo/citologia , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
4.
Front Neuroendocrinol ; 32(1): 95-107, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21129392

RESUMO

Fibroblast growth factor (FGF) signaling is pivotal to the formation of numerous central regions. Increasing evidence suggests FGF signaling also directs the development of the neuroendocrine hypothalamus, a collection of neuroendocrine neurons originating primarily within the nose and the ventricular zone of the diencephalon. This review outlines evidence for a role of FGF signaling in the prenatal and postnatal development of several hypothalamic neuroendocrine systems. The emphasis is placed on the nasally derived gonadotropin-releasing hormone neurons, which depend on neurotrophic cues from FGF signaling throughout the neurons' lifetime. Although less is known about neuroendocrine neurons derived from the diencephalon, recent studies suggest they also exhibit variable levels of dependence on FGF signaling. Overall, FGF signaling provides a broad spectrum of cues that ranges from genesis, cell survival/death, migration, morphological changes, to hormone synthesis in the neuroendocrine hypothalamus. Abnormal FGF signaling will deleteriously impact multiple hypothalamic neuroendocrine systems, resulting in the disruption of diverse physiological functions.


Assuntos
Fatores de Crescimento de Fibroblastos/fisiologia , Hipotálamo/embriologia , Sistemas Neurossecretores/embriologia , Animais , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hipotálamo/metabolismo , Modelos Biológicos , Sistemas Neurossecretores/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
5.
Biol Reprod ; 86(4): 119, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22278983

RESUMO

Fibroblast growth factor (FGF) signaling is essential for the development of the gonadotropin-releasing hormone (GnRH) system. Mice harboring deficiencies in Fgf8 or Fgf receptor 1 (Fgfr1) suffer a significant loss of GnRH neurons, but their reproductive phenotypes have not been examined. This study examined if female mice hypomorphic for Fgf8, Fgfr1, or both (compound hypomorphs) exhibited altered parameters of pubertal onset, estrous cyclicity, and fertility. Further, we examined the number of kisspeptin (KP)-immunoreactive (ir) neurons in the anteroventral periventricular/periventricular nuclei (AVPV/PeV) of these mice to assess if changes in the KP system, which stimulates the GnRH system, could contribute to the reproductive phenotypes. Single hypomorphs (Fgfr1(+/-) or Fgf8(+/-)) had normal timing for vaginal opening (VO) but delayed first estrus. However, after achieving the first estrus, they underwent normal expression of estrous cycles. In contrast, the compound hypomorphs underwent early VO and normal first estrus, but had disorganized estrous cycles that subsequently reduced their fertility. KP immunohistochemistry on Postnatal Day 15, 30, and 60 transgenic female mice revealed that female compound hypomorphs had significantly more KP-ir neurons in the AVPV/PeV compared to their wild-type littermates, suggesting increased KP-ir neurons may drive early VO but could not maintain the cyclic changes in GnRH neuronal activity required for female fertility. Overall, these data suggest that Fgf signaling deficiencies differentially alter the parameters of female pubertal onset and cyclicity. Further, these deficiencies led to changes in the AVPV/PeV KP-ir neurons that may have contributed to the accelerated VO in the compound hypomorphs.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Infertilidade Feminina/metabolismo , Kisspeptinas/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Reprodução/fisiologia , Animais , Núcleos Anteriores do Tálamo/metabolismo , Comunicação Celular/fisiologia , Ciclo Estral/metabolismo , Feminino , Fator 8 de Crescimento de Fibroblasto/genética , Hormônio Liberador de Gonadotropina/metabolismo , Camundongos , Camundongos Transgênicos , Núcleos da Linha Média do Tálamo/metabolismo , Neurônios/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Maturidade Sexual/fisiologia , Transdução de Sinais/fisiologia
6.
J Clin Invest ; 118(8): 2822-31, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18596921

RESUMO

Idiopathic hypogonadotropic hypogonadism (IHH) with anosmia (Kallmann syndrome; KS) or with a normal sense of smell (normosmic IHH; nIHH) are heterogeneous genetic disorders associated with deficiency of gonadotropin-releasing hormone (GnRH). While loss-of-function mutations in FGF receptor 1 (FGFR1) cause human GnRH deficiency, to date no specific ligand for FGFR1 has been identified in GnRH neuron ontogeny. Using a candidate gene approach, we identified 6 missense mutations in FGF8 in IHH probands with variable olfactory phenotypes. These patients exhibited varied degrees of GnRH deficiency, including the rare adult-onset form of hypogonadotropic hypogonadism. Four mutations affected all 4 FGF8 splice isoforms (FGF8a, FGF8b, FGF8e, and FGF8f), while 2 mutations affected FGF8e and FGF8f isoforms only. The mutant FGF8b and FGF8f ligands exhibited decreased biological activity in vitro. Furthermore, mice homozygous for a hypomorphic Fgf8 allele lacked GnRH neurons in the hypothalamus, while heterozygous mice showed substantial decreases in the number of GnRH neurons and hypothalamic GnRH peptide concentration. In conclusion, we identified FGF8 as a gene implicated in GnRH deficiency in both humans and mice and demonstrated an exquisite sensitivity of GnRH neuron development to reductions in FGF8 signaling.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Hormônio Liberador de Gonadotropina/deficiência , Transdução de Sinais , Adulto , Animais , Estudos de Casos e Controles , Estudos de Coortes , Feminino , Fator 8 de Crescimento de Fibroblasto/química , Fator 8 de Crescimento de Fibroblasto/genética , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Heterozigoto , Humanos , Hipogonadismo/genética , Hipogonadismo/fisiopatologia , Síndrome de Kallmann/genética , Síndrome de Kallmann/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Mutação , Neurônios/citologia , Neurônios/metabolismo , Transtornos do Olfato/genética , Linhagem
7.
Endocrinology ; 162(9)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34147032

RESUMO

The rapid decline of circulating 17ß-estradiol (E2) at menopause leads to negative neurological consequences, although hormone therapy paradoxically has both harmful and positive effects depending on the age at which it is delivered. The inconsistent response to E2 suggests unappreciated regulatory mechanisms for estrogen receptors (ERs), and we predicted it could be due to age-related differences in ERß phosphorylation. We assessed ERß phosphorylation using a sensitive mass spectrometry approach that provides absolute quantification (AQUA-MS) of individually phosphorylated residues. Specifically, we quantified phosphorylated ERß in the hippocampus of women (aged 21-83 years) and in a rat model of menopause at 4 residues with conserved sequence homology between the 2 species: S105, S176, S200, and Y488. Phosphorylation at these sites, which spanned all domains of ERß, were remarkably consistent between the 2 species, showing high levels of S105 phosphorylation (80%-100%) and low levels of S200 (20%-40%). Further, S200 phosphorylation decreased with aging in humans and loss of E2 in rats. Surprisingly, Y488 phosphorylation, which has been linked to ERß ligand-independent actions, exhibited approximately 70% phosphorylation, unaltered by species, age, or E2, suggesting ERß's primary mode of action may not require E2 binding. We further show phosphorylation at 2 sites directly altered ERß DNA-binding efficiency, and thus could affect its transcription factor activity. These findings provide the first absolute quantification of ERß phosphorylation in the human and rat brain, novel insights into ERß regulation, and a critical foundation for providing more targeted therapeutic options for menopause in the future.


Assuntos
Receptor beta de Estrogênio/análise , Hipocampo/química , Menopausa/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/metabolismo , Envelhecimento/patologia , Aminoácidos/análise , Aminoácidos/metabolismo , Animais , Estradiol/análise , Estradiol/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Pessoa de Meia-Idade , Modelos Animais , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/metabolismo , Fosforilação , Ratos , Ratos Endogâmicos F344 , Adulto Jovem
8.
J Neuroendocrinol ; 32(6): e12860, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32452569

RESUMO

Mammalian reproductive success depends on gonadotrophin-releasing hormone (GnRH) neurones to stimulate gonadotrophin secretion from the anterior pituitary and activate gonadal steroidogenesis and gametogenesis. Genetic screening studies in patients diagnosed with Kallmann syndrome (KS), a congenital form of hypogonadotrophic hypogonadism (CHH), identified several causal mutations, including those in the fibroblast growth factor (FGF) system. This signalling pathway regulates neuroendocrine progenitor cell proliferation, fate specification and cell survival. Indeed, the GnRH neurone system was absent or abrogated in transgenic mice with reduced (ie, hypomorphic) Fgf8 and/or Fgf receptor (Fgfr) 1 expression, respectively. Moreover, we found that GnRH neurones were absent in the embryonic olfactory placode of Fgf8 hypomorphic mice, the putative birthplace of GnRH neurones. These observations, together with those made in human KS/CHH patients, indicate that the FGF8/FGFR1 signalling system is a requirement for the ontogenesis of the GnRH neuronal system and function. In this review, we discuss how epigenetic factors control the expression of genes such as Fgf8 that are known to be critical for GnRH neurone ontogenesis, fate specification, and the pathogenesis of KS/CHH.


Assuntos
Epigênese Genética/fisiologia , Hipogonadismo/genética , Neurogênese/genética , Neurônios/fisiologia , Animais , Epigenômica , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Hipogonadismo/patologia , Hipogonadismo/psicologia , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/patologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/fisiologia
9.
Neurosci Lett ; 714: 134569, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31644920

RESUMO

Down syndrome is the most common genetic cause of intellectual disability and occurs due to the trisomy of human chromosome 21. Adolescent and adult brains from humans with Down syndrome exhibit various neurological phenotypes including a reduction in the size of the corpus callosum, hippocampal commissure and anterior commissure. However, it is unclear when and how these interhemispheric connectivity defects arise. Using the Ts65Dn mouse model of Down syndrome, we examined interhemispheric connectivity in postnatal day 0 (P0) Ts65Dn mouse brains. We find that there is no change in the volume of the corpus callosum or anterior commissure in P0 Ts65Dn mice. However, the volume of the hippocampal commissure is significantly reduced in P0 Ts65Dn mice, and this may contribute to the impaired learning and memory phenotype of this disorder. Interhemispheric connectivity defects that arise during development may be due to disrupted axon growth. In line with this, we find that developing hippocampal neurons display reduced axon length in vitro, as compared to neurons from their euploid littermates. This study is the first to report the presence of defective interhemispheric connectivity at the time of birth in Ts65Dn mice, providing evidence that early therapeutic intervention may be an effective time window for the treatment of Down syndrome.


Assuntos
Comissura Anterior/patologia , Axônios/patologia , Corpo Caloso/patologia , Síndrome de Down/patologia , Fórnice/patologia , Animais , Animais Recém-Nascidos , Comissura Anterior/fisiopatologia , Orientação de Axônios/fisiologia , Tamanho Celular , Corpo Caloso/fisiopatologia , Modelos Animais de Doenças , Síndrome de Down/fisiopatologia , Fórnice/fisiopatologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Vias Neurais , Neurogênese/fisiologia , Crescimento Neuronal , Neurônios/patologia , Tamanho do Órgão
10.
PLoS One ; 14(7): e0220530, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31361780

RESUMO

Fibroblast growth factor 8 (FGF8) is a potent morphogen that regulates the ontogenesis of gonadotropin-releasing hormone (GnRH) neurons, which control the hypothalamus-pituitary-gonadal (HPG) axis, and therefore reproductive success. Indeed, FGF8 and FGFR1 deficiency severely compromises vertebrate reproduction in mice and humans and is associated with Kallmann Syndrome (KS), a congenital disease characterized by hypogonadotropic hypogonadism associated with anosmia. Our laboratory demonstrated that FGF8 signaling through FGFR1, both of which are KS-related genes, is necessary for proper GnRH neuron development in mice and humans. Here, we investigated the possibility that non-genetic factors, such as the epigenome, may contribute to KS onset. For this purpose, we developed an embryonic explant model, utilizing the mouse olfactory placode (OP), the birthplace of GnRH neurons. We show that TET1, which converts 5-methylcytosine residues (5mC) to 5-hydroxymethylated cytosines (5hmC), controls transcription of Fgf8 during GnRH neuron ontogenesis. Through MeDIP and ChIP RT-qPCR we found that TET1 bound to specific CpG islands on the Fgf8 promoter. We found that the temporal expression of Fgf8 correlates with not only TET1 binding, but also with 5hmC enrichment. siRNA knockdown of Tet1 reduced Fgf8 and Fgfr1 mRNA expression. During this time period, Fgf8 also switched histone status, most likely via recruitment of EZH2, a major component of the polycomb repressor complex-2 (PRC2) at E13.5. Together, these studies underscore the significance of epigenetics and chromatin modifications to temporally regulated genes involved in KS.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fator 8 de Crescimento de Fibroblasto/genética , Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transcrição Gênica , Animais , Células Cultivadas , Proteínas de Ligação a DNA/genética , Feminino , Histonas/genética , Histonas/metabolismo , Masculino , Camundongos , Neurônios/citologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais
11.
Endocrinology ; 149(10): 4997-5003, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18566132

RESUMO

GnRH neurons are essential for the onset and maintenance of reproduction. Mutations in both fibroblast growth factor receptor (Fgfr1) and Fgf8 have been shown to cause Kallmann syndrome, a disease characterized by hypogonadotropic hypogonadism and anosmia, indicating that FGF signaling is indispensable for the formation of a functional GnRH system. Presently it is unclear which stage of GnRH neuronal development is most impacted by FGF signaling deficiency. GnRH neurons express both FGFR1 and -3; thus, it is also unclear whether FGFR1 or FGFR3 contributes directly to GnRH system development. In this study, we examined the developing GnRH system in mice deficient in FGF8, FGFR1, or FGFR3 to elucidate the individual contribution of these FGF signaling components. Our results show that the early emergence of GnRH neurons from the embryonic olfactory placode requires FGF8 signaling, which is mediated through FGFR1, not FGFR3. These data provide compelling evidence that the developing GnRH system is exquisitely sensitive to reduced levels of FGF signaling. Furthermore, Kallmann syndrome stemming from FGF signaling deficiency may be due primarily to defects in early GnRH neuronal development prior to their migration into the forebrain.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Hormônio Liberador de Gonadotropina/fisiologia , Condutos Olfatórios/embriologia , Condutos Olfatórios/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/fisiologia , Animais , Apoptose/fisiologia , Movimento Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Hipotálamo/citologia , Hipotálamo/embriologia , Hipotálamo/fisiologia , Proteínas de Filamentos Intermediários/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Condutos Olfatórios/citologia , Periferinas , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Transativadores/metabolismo , Órgão Vomeronasal/citologia , Órgão Vomeronasal/embriologia , Órgão Vomeronasal/fisiologia
12.
Endocrinology ; 148(7): 3371-82, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17412808

RESUMO

Arginine vasopressin (AVP) is a neuropeptide involved in the regulation of fluid balance, stress, circadian rhythms, and social behaviors. In the brain, AVP is tightly regulated by gonadal steroid hormones in discrete regions with gonadectomy abolishing and testosterone replacement restoring normal AVP expression in adult males. Previous studies demonstrated that 17beta-estradiol, a primary metabolite of testosterone, is responsible for restoring most of the AVP expression in the brain after castration. However, 5alpha-dihydrotestosterone (DHT) has also been shown to play a role in the regulation of AVP expression, thus implicating the involvement of both androgen and estrogen receptors (ER). Furthermore, DHT, through its conversion to 5alpha-androstane-3beta,17beta-diol, has been shown to modulate estrogen response element-mediated promoter activity through an ER pathway. The present study addressed two central hypotheses: 1) that androgens directly modulate AVP promoter activity and 2) the effect is mediated by an estrogen or androgen receptor pathway. To that end, we overexpressed androgen receptor, ERbeta, and ERbeta splice variants in a neuronal cell line and measured AVP promoter activity using a firefly luciferase reporter assay. Our results demonstrate that DHT and its metabolite 5alpha-androstane-3beta,17beta-diol stimulate AVP promoter activity through ERbeta in a neuronal cell line.


Assuntos
Arginina Vasopressina/genética , Di-Hidrotestosterona/farmacologia , Receptor beta de Estrogênio/genética , Neurônios/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Androgênios/química , Androgênios/metabolismo , Androstano-3,17-diol/farmacologia , Arginina Vasopressina/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Receptor beta de Estrogênio/metabolismo , Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/genética , Humanos , Imuno-Histoquímica , Luciferases/genética , Luciferases/metabolismo , Dados de Sequência Molecular , Estrutura Molecular , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Comp Neurol ; 505(3): 249-67, 2007 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-17879269

RESUMO

Estrogens regulate neural processes such as neuronal development, reproductive behavior, and hormone secretion, and signal through estrogen receptor (ER) alpha and ERbeta (here called ERbeta1). Recent studies have found variations in ERalpha and ERbeta1 mRNA splicing in rodents and humans. Functional reporter gene assays suggest that these splicing variations alter ER-mediated transcriptional regulation. Estrogen receptor beta 2 (ERbeta2), an ERbeta1 splice variant containing an 18 amino acid (AA) insert in the ligand binding domain, binds estradiol with approximately 10-fold lower affinity than ERbeta1, suggesting that it may serve as a low-affinity ER. Moreover, ERbeta2 reportedly acts in a dominant-negative fashion when heterodimerized with ERbeta1 or ERalpha. To explore the function of ERbeta2 in brain, an antiserum (TwobetaER.1) targeting the 18 AA insert was developed and characterized. Western blot analysis and transient expression of ERbeta2 in cell lines demonstrated that TwobetaER.1 recognizes ERbeta2. In the adult female rat brain, ERbeta2 immunoreactivity is localized in the cell nucleus and is expressed with a distribution similar to that of ERbeta1 mRNA. ERbeta2 immunoreactive cell numbers were high in, for example, piriform cortex, paraventricular nucleus, supraoptic nucleus, arcuate nucleus, and hippocampal CA regions, whereas it was low in the dentate gyrus. Moreover, ERbeta2 is coexpressed in gonadotropin-releasing hormone and oxytocin neurons. These studies demonstrate ERbeta splice variant proteins in brain and support the hypothesis that ER signaling diversity depends not only on ligand or coregulatory proteins, but also on regional and phenotypic selectivity of ER splice variant proteins.


Assuntos
Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Mesencéfalo/metabolismo , Prosencéfalo/metabolismo , Processamento Alternativo , Animais , Western Blotting , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Ovariectomia , Ocitocina/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
14.
Endocrinology ; 147(4): 1924-31, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16439454

RESUMO

GnRH is the most upstream regulator of reproduction in vertebrates, and its synthesis and release are regulated by gonadal steroid hormones. The proposed sites of hormone action were historically thought to be upstream from GnRH neurons; however, the discovery of ERbeta in a subset of GnRH neurons suggests that this hypothesis should be reevaluated. To determine a functional role for ERbeta in GnRH neurons, we examined ERbeta's regulation of GnRH promoter activity. The GnRH-producing cell line, GT1-7, was cotransfected with expression vectors containing one of three ERbeta splice variants and a luciferase-reporter construct containing the full-length mouse GnRH promoter sequence or one of two deletions upstream of the transcription start site (-225/-201; -184/-150). Transfected cells were treated with 100 nm 17beta-estradiol (E(2)), diarylpropionitrile, raloxifene, or vehicle. There was a robust increase in GnRH-luciferase activity by all ERbeta splice variants in the absence of hormone. Furthermore, E(2) treatment abolished this response for ER-beta1 and ER-beta2, but not ER-beta1delta3. The -225/-201 and -184/-150 regions were critical for ERbeta-induced promoter activity because deletion of these regions eliminated the ligand-independent effects of ERbeta. ER-beta1 binds directly to these promoter regions and because there are no classical estrogen response elements in the mouse GnRH promoter, these data raise the possibility that this region contains a novel estrogen response element specific for ERbeta. Overall, our data suggest that ERbeta functions as a basic transcription factor in GnRH neurons and demonstrate a potential molecular mechanism for the negative feedback effects of E(2) on GnRH.


Assuntos
Receptor beta de Estrogênio/fisiologia , Hormônio Liberador de Gonadotropina/genética , Regiões Promotoras Genéticas , Processamento Alternativo , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular , AMP Cíclico/biossíntese , Receptor beta de Estrogênio/genética , Retroalimentação Fisiológica , Ligantes , Camundongos , Dados de Sequência Molecular
15.
Brain Res ; 1082(1): 50-60, 2006 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-16513095

RESUMO

Perinatal 17beta-estradiol (E2) rapidly and markedly affects the morphological and neurochemical organization of the vertebrate brain. For instance, the sex difference in perinatal progestin receptor (PR) immunoreactivity in the medial preoptic nucleus (MPN) of the rat brain is due to the intracellular conversion of testosterone into E2 in males. Neonatal alpha-fetoprotein prevents circulating estrogens from accessing the brain, therefore, to overcome alpha-fetoprotein sequestration of E2, estrogen replacement studies during development have used natural and synthetic estrogen dosages in the milligram to microgram range. These levels could be considered as supraphysiological. Moreover, it is not clear through which ER subtype E2 acts to induce PR expression in the neonatal rat MPN because E2 binds similarly to estrogen receptor (ER)alpha and ERbeta. Consequently, we investigated whether nanogram levels of E2 affected PR protein and mRNA levels in the neonatal MPN. Furthermore, propylpyrazole-triol (PPT), a highly selective agonist for ERalpha, and diarylpropionitrile (DPN), a highly selective agonist for ERbeta, were used to determine if E2-dependent PR expression in the neonatal rat is mediated through ERalpha and/or ERbeta. Immunocytochemistry and quantitative real-time RT-PCR determined that as little as 100 ng E2 significantly induced PR protein and mRNA in the female and neonatally castrated male MPN on PN 4, indicating that the neonatal rat brain is highly sensitive to circulating estrogens. PPT, but not DPN, induced PR expression in the neonatal MPN and arcuate nucleus (Arc), demonstrating that PR expression in the neonatal rat brain depends solely on E2 activated ERalpha. In the lateral bed nucleus of the stria terminalis (BSTL), neither PPT nor DPN affected PR expression, suggesting the presence of a gonadal hormone-independent PR regulatory mechanism.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Receptores de Progesterona/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Castração/métodos , Contagem de Células/métodos , Diagnóstico por Imagem/métodos , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Imuno-Histoquímica/métodos , Masculino , Nitrilas/farmacologia , Fenóis , Gravidez , Propionatos/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Pirazóis/farmacologia , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores de Progesterona/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Caracteres Sexuais
16.
Front Cell Dev Biol ; 4: 34, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27200347

RESUMO

Fibroblast growth factor 8 (FGF8) is a potent morphogen that regulates the embryonic development of hypothalamic neuroendocrine cells. Indeed, using Fgf8 hypomorphic mice, we showed that reduced Fgf8 mRNA expression completely eliminated the presence of gonadotropin-releasing hormone (GnRH) neurons. These findings suggest that FGF8 signaling is required during the embryonic development of mouse GnRH neurons. Additionally, in situ hybridization studies showed that the embryonic primordial birth place of GnRH neurons, the olfactory placode, is highly enriched for Fgf8 mRNA expression. Taken together these data underscore the importance of FGF8 signaling for GnRH emergence. However, an important question remains unanswered: How is Fgf8 gene expression regulated in the developing embryonic mouse brain? One major candidate is the androgen receptor (AR), which has been shown to upregulate Fgf8 mRNA in 60-70% of newly diagnosed prostate cancers. Therefore, we hypothesized that ARs may be involved in the regulation of Fgf8 transcription in the developing mouse brain. To test this hypothesis, we used chromatin-immunoprecipitation (ChIP) assays to elucidate whether ARs interact with the 5'UTR region upstream of the translational start site of the Fgf8 gene in immortalized mouse GnRH neurons (GT1-7) and nasal explants. Our data showed that while AR interacts with the Fgf8 promoter region, this interaction was androgen-independent, and that androgen treatment did not affect Fgf8 mRNA levels, indicating that androgen signaling does not induce Fgf8 transcription. In contrast, inhibition of DNA methyltransferases (DNMT) significantly upregulated Fgf8 mRNA levels indicating that Fgf8 transcriptional activity may be dependent on DNA methylation status.

17.
Artigo em Inglês | MEDLINE | ID: mdl-27656162

RESUMO

Over the last few years, numerous studies solidified the hypothesis that fibroblast growth factor (FGF) signaling regulates neuroendocrine progenitor cell proliferation, fate specification, and cell survival and, therefore, is critical for the regulation and maintenance of homeostasis of the body. One important example that underscores the involvement of FGF signaling during neuroendocrine cell development is gonadotropin-releasing hormone (GnRH) neuron ontogenesis. Indeed, transgenic mice with reduced olfactory placode (OP) Fgf8 expression do not have GnRH neurons. This observation indicates the requirement of FGF8 signaling for the emergence of the GnRH neuronal system in the embryonic OP, the putative birth place of GnRH neurons. Mammalian reproductive success depends on the presence of GnRH neurons to stimulate gonadotropin secretion from the anterior pituitary, which activates gonadal steroidogenesis and gametogenesis. Together, these observations are critical for understanding the function of GnRH neurons and their control of the hypothalamus-pituitary-gonadal (HPG) axis to maintain fertility. Taken together, these studies illustrate that GnRH neuron emergence and hence HPG function is vulnerable to genomic and molecular signals that abnormally modify Fgf8 expression in the developing mouse OP. In this short review, we focus on research that is aimed at unraveling how androgen, all-trans retinoic acid, and how epigenetic factors modify control mouse OP Fgf8 transcription in the context of GnRH neuronal development and mammalian reproductive success.

18.
Brain Res ; 1646: 287-296, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27291295

RESUMO

Our previous studies showed that Fgf8 mutations can cause Kallmann syndrome (KS), a form of congenital hypogonadotropic hypogonadism, in which patients do not undergo puberty and are infertile. Interestingly, some KS patients also have agenesis of the corpus callosum (ACC) suggesting that KS pathology is not limited to reproductive function. Here, we asked whether FGF8 dysfunction is the underlying cause of ACC in some KS patients. Indeed, early studies in transgenic mice with Fgf8 mutations reported the presence of failed or incomplete corpus callosum formation. Additional studies in transgenic mice showed that FGF8 function most likely prevents the prenatal elimination of glial fibrillary acidic protein (GFAP)-immunoreactive (IR) glial cells in the indusium griseum (IG) and midline zipper (MZ), two anterior-dorsal midline regions required for corpus callosum formation (i.e., between embryonic days (E) 15.5-18.5). Here, we tested the hypothesis that FGF8 function is critical for the survival of the GFAP-IR midline glial cells. First, we measured the incidence of apoptosis in the anterior-dorsal midline region in Fgf8 hypomorphic mice during embryonic corpus callosum formation. Second, we quantified the GFAP expression in the anterior-dorsal midbrain region during pre- and postnatal development, in order to study: 1) how Fgf8 hypomorphy disrupts prenatal GFAP-IR midline glial cell development, and 2) whether Fgf8 hypomorphy continues to disrupt postnatal GFAP-IR midline glial cell development. Our results indicate that perinatal FGF8 signaling is important for the timing of the onset of anterior-dorsal Gfap expression in midline glial cells suggesting that FGF8 function regulates midline GFAP-IR glial cell development, which when disrupted by Fgf8 deficiency prevents the formation of the corpus callosum. These studies provide an experimentally-based mechanistic explanation as to why corpus callosum formation may fail in KS patients with deficits in FGF signaling.


Assuntos
Astrócitos/fisiologia , Corpo Caloso/embriologia , Fator 8 de Crescimento de Fibroblasto/fisiologia , Síndrome de Kallmann/patologia , Animais , Apoptose , Astrócitos/citologia , Astrócitos/metabolismo , Corpo Caloso/citologia , Corpo Caloso/patologia , Transportador 1 de Aminoácido Excitatório/metabolismo , Feminino , Fator 8 de Crescimento de Fibroblasto/genética , Proteína Glial Fibrilar Ácida/metabolismo , Síndrome de Kallmann/embriologia , Masculino , Camundongos , Camundongos Transgênicos
19.
J Neurosci ; 22(3): 1027-33, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11826131

RESUMO

Gonadal steroids have remarkable developmental effects on sex-dependent brain organization and behavior in animals. Presumably, fetal or neonatal gonadal steroids are also responsible for sexual differentiation of the human brain. A limbic structure of special interest in this regard is the sexually dimorphic central subdivision of the bed nucleus of the stria terminalis (BSTc), because its size has been related to the gender identity disorder transsexuality. To determine at what age the BSTc becomes sexually dimorphic, the BSTc volume in males and females was studied from midgestation into adulthood. Using vasoactive intestinal polypeptide and somatostatin immunocytochemical staining as markers, we found that the BSTc was larger and contains more neurons in men than in women. However, this difference became significant only in adulthood, showing that sexual differentiation of the human brain may extend into the adulthood. The unexpectedly late sexual differentiation of the BSTc is discussed in relation to sex differences in developmental, adolescent, and adult gonadal steroid levels.


Assuntos
Neurônios/citologia , Núcleos Septais/anatomia & histologia , Núcleos Septais/fisiologia , Caracteres Sexuais , Diferenciação Sexual/fisiologia , Adolescente , Adulto , Contagem de Células , Diferenciação Celular/fisiologia , Criança , Pré-Escolar , Feminino , Humanos , Imuno-Histoquímica , Lactente , Masculino , Pessoa de Meia-Idade , Neurônios/metabolismo , Núcleos Septais/embriologia , Fatores Sexuais , Maturidade Sexual/fisiologia , Somatostatina/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo
20.
Endocrinology ; 146(2): 797-807, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15514081

RESUMO

Estrogens are reported to have both anxiogenic and anxiolytic properties. This dichotomous neurobiological response to estrogens may be mediated by the existence of two distinct estrogen receptor (ER) systems, ERalpha and ERbeta. In brain, ERalpha plays a critical role in regulating reproductive neuroendocrine function, whereas ERbeta may be more important in regulating nonreproductive functions. To determine whether estrogen's anxiolytic actions could be mediated by ERbeta, we examined anxiety-related behaviors after treatment with ER subtype-selective agonists. Ovariectomized female rats, divided into four treatment groups, were injected with the selective ERbeta agonist diarylpropionitrile (DPN), the ERalpha-selective agonist propyl-pyrazole-triol (PPT), 17beta-estradiol, or vehicle daily for 4d. After injections, behavior was monitored in the elevated plus maze or open field. Rats treated with DPN showed significantly decreased anxiety-related behaviors in both behavioral paradigms. In the elevated plus maze, DPN significantly increased the number of open arm entries and time spent on the open arms of the maze. Furthermore, DPN significantly reduced, whereas PPT increased, anxiogenic behaviors such as the number of fecal boli and time spent grooming. In the open field, DPN-treated females made more rears, interacted more with a novel object, and spent more time in the middle of the open field than did control or PPT-treated rats. To confirm that DPN's anxiolytic actions are ER mediated, the nonselective ER antagonist tamoxifen was administered alone or in combination with DPN. Tamoxifen blocked the previously identified anxiolytic actions of DPN. Taken together, these findings suggest that the anxiolytic properties of estrogens are ERbeta mediated.


Assuntos
Ansiedade/tratamento farmacológico , Ansiedade/metabolismo , Encéfalo/metabolismo , Receptor beta de Estrogênio/metabolismo , Animais , Comportamento Animal/fisiologia , Ligação Competitiva , Barreira Hematoencefálica , Encéfalo/efeitos dos fármacos , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/agonistas , Medo/efeitos dos fármacos , Feminino , Masculino , Nitrilas/metabolismo , Nitrilas/farmacocinética , Fenóis , Propionatos/metabolismo , Propionatos/farmacocinética , Pirazóis/metabolismo , Pirazóis/farmacocinética , Ratos , Ratos Sprague-Dawley , Receptores de Progesterona/metabolismo , Regulação para Cima/efeitos dos fármacos , Útero/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA