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.
Int J Obes (Lond) ; 41(1): 149-158, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27773938

RESUMO

BACKGROUND: Consumption of dietary fat is one of the key factors leading to obesity. High-fat diet (HFD)-induced obesity is characterized by induction of inflammation in the hypothalamus; however, the temporal regulation of proinflammatory markers and their impact on hypothalamic appetite-regulating neuropeptide Y/agouti-related peptide (NPY/AgRP) neurons remains undefined. METHODS: Mice were injected with an acute lipid infusion for 24 h or fed a HFD over 8-20 weeks. Characterized mouse NPY/AgRP hypothalamic cell lines were used for in vitro experimentation. Immunohistochemistry in brain slices or quantitative real-time PCR in cell lines, was performed to determine changes in the expression of key inflammatory markers and neuropeptides. RESULTS: Hypothalamic inflammation, indicated by tumor necrosis factor (TNF)-α expression and astrocytosis in the arcuate nucleus, was evident following acute lipid infusion. HFD for 8 weeks suppressed TNF-α, while significantly increasing heat-shock protein 70 and ciliary neurotrophic factor, both neuroprotective components. HFD for 20 weeks induced TNF-α expression in NPY/AgRP neurons, suggesting a detrimental temporal regulatory mechanism. Using NPY/AgRP hypothalamic cell lines, we found that palmitate provoked a mixed inflammatory response on a panel of inflammatory and endoplasmic reticulum (ER) stress genes, whereas TNF-α significantly upregulated IκBα, nuclear factor (NF)-κB and interleukin-6 mRNA levels. Palmitate and TNF-α exposure predominantly induced NPY mRNA levels. Utilizing an I kappa B kinase ß (IKKß) inhibitor, we demonstrated that these effects potentially occur via the inflammatory IKKß/NF-κB pathway. CONCLUSIONS: These findings indicate that acute lipid and chronic HFD feeding in vivo, as well as acute palmitate and TNF-α exposure in vitro, induce markers of inflammation or ER stress in the hypothalamic appetite-stimulating NPY/AgRP neurons over time, which may contribute to a dramatic alteration in NPY/AgRP content or expression. Acute and chronic HFD feeding in vivo temporally regulates arcuate TNF-α expression with reactive astrocytosis, which suggests a time-dependent neurotrophic or neurotoxic role of lipids.


Assuntos
Apetite/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Hipotálamo/patologia , Inflamação/induzido quimicamente , Neurônios/efeitos dos fármacos , Neuropeptídeo Y/metabolismo , Palmitatos/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Núcleo Arqueado do Hipotálamo/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Hipotálamo/efeitos dos fármacos , Inflamação/patologia , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Obesidade/patologia
2.
J Neuroendocrinol ; 26(12): 888-97, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25283492

RESUMO

RF-amide-related peptide-3 [RFRP-3; also often referred to as the mammalian orthologue of the avian gonadotrophin-inhibitory hormone (GnIH)] and kisspeptin have emerged as potent modulators of neuroendocrine function via direct regulation of the reproductive axis in the hypothalamus and pituitary. There are few studies focusing on the direct regulatory effects of RFRP-3 and kisspeptin on gonadotrophin-releasing hormones (GnRH) neurones. We report their effect on GnRH mRNA expression and release in a novel GnRH neuronal cell model, mHypoA-GnRH/GFP, generated from adult-derived GnRH-GFP neurones. The neurones express receptors for both RFRP-3 and kisspeptin, Gpr147 and Gpr54, respectively. Incubation with 100 nm RFRP-3 results in attenuation of GnRH mRNA expression by approximately 60%. Conversely, incubation with 10 nm of Kiss-10 induced GnRH mRNA expression, whereas the combined effect was an overall repression of GnRH mRNA levels. With transcription inhibitors, the repression of GnRH mRNA levels was linked to a transcriptional mechanism but not mRNA stability. No significant changes in GnRH secretion were observed upon RFRP-3 exposure in these neurones. Our findings suggest that the suppressive signalling of RFRP-3 on GnRH transcription may dominate over kisspeptin induction in the mHypoA-GnRH/GFP GnRH neuronal cell model.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Kisspeptinas/farmacologia , Neuropeptídeos/farmacologia , Transcrição Gênica/efeitos dos fármacos , Animais , Benzimidazóis/farmacologia , Linhagem Celular , Dactinomicina/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/antagonistas & inibidores , Kisspeptinas/metabolismo , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Receptores de Neuropeptídeos/metabolismo
3.
Diabetologia ; 56(5): 1057-67, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23475366

RESUMO

AIMS/HYPOTHESIS: Endoplasmic reticulum (ER) stress has been detected in pancreatic beta cells and in insulin-sensitive tissues, such as adipose and liver, in obesity-linked rodent models of type 2 diabetes. The contribution of ER stress to pancreatic beta cell dysfunction in type 2 diabetes is unclear. We hypothesised that increased chaperone capacity protects beta cells from ER stress and dysfunction caused by obesity and improves overall glucose homeostasis. METHODS: We generated a mouse model that overproduces the resident ER chaperone GRP78 (glucose-regulated protein 78 kDa) in pancreatic beta cells under the control of a rat insulin promoter. These mice were subjected to high-fat diet (HFD) feeding for 20 weeks and metabolic variables and markers of ER stress in islets were measured. RESULTS: As expected, control mice on the HFD developed obesity, glucose intolerance and insulin resistance. In contrast, GRP78 transgenic mice tended to be leaner than their non-transgenic littermates and were protected against development of glucose intolerance, insulin resistance and ER stress in islets. Furthermore, islets from transgenic mice had a normal insulin content and normal levels of cell-surface GLUT2 (glucose transporter 2) and the transgenic mice were less hyperinsulinaemic than control mice on the HFD. CONCLUSIONS/INTERPRETATION: These data show that increased chaperone capacity in beta cells provides protection against the pathogenesis of obesity-induced type 2 diabetes by maintaining pancreatic beta cell function, which ultimately improves whole-body glucose homeostasis.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Estresse do Retículo Endoplasmático , Proteínas de Choque Térmico/biossíntese , Células Secretoras de Insulina/metabolismo , Regulação para Cima , Animais , Cruzamentos Genéticos , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/patologia , Dieta Hiperlipídica/efeitos adversos , Chaperona BiP do Retículo Endoplasmático , Transportador de Glucose Tipo 2/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Hiperinsulinismo/complicações , Hiperinsulinismo/prevenção & controle , Insulina/genética , Insulina/metabolismo , Resistência à Insulina , Secreção de Insulina , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/complicações , Obesidade/prevenção & controle , Regiões Promotoras Genéticas , Ratos , Propriedades de Superfície , Técnicas de Cultura de Tecidos
4.
Neuroscience ; 219: 255-70, 2012 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-22698694

RESUMO

The pyramidal neurons in the hippocampus are extremely neuroplastic, and the complexity of dendritic branches can be dynamically altered in response to a variety of stimuli, including learning and stress. Recently, the teneurin family of proteins has emerged as an interneuronal and extracellular matrix signaling system that plays a significant role in brain development and neuronal communication. Encoded on the last exon of the teneurin genes is a new family of bioactive peptides termed the teneurin C-terminal-associated peptides (TCAPs). Previous studies indicate that TCAP-1 regulates axon fasciculation and dendritic morphology in the hippocampus. This study was aimed at understanding the molecular mechanisms by which TCAP-1 regulates these changes in the mouse hippocampus. Fluoresceinisothiocyanate (FITC)-labeled TCAP-1 binds to the pyramidal neurons of the CA2 and CA3, and dentate gyrus in the hippocampus of the mouse brain. Moreover, FITC-TCAP-1 co-localizes with ß-dystroglycan upon binding to the plasma membrane of cultured immortalized mouse E14 hippocampal cells. In culture, TCAP-1 stimulates ERK1/2-dependent phosphorylation of the cytoskeletal regulatory proteins, stathmin at serine-25 and filamin A at serine-2152. In addition, TCAP-1 induces actin polymerization, increases immunoreactivity of tubulin-based cytoskeletal elements and causes a corresponding increase in filopodia formation and mean filopodia length in cultured hippocampal cells. We postulate that the TCAP-1 region of teneurin-1 has a direct action on the cytoskeletal reorganization that precedes neurite and process development in hippocampal neurons. Our data provides novel evidence that functionally links the teneurin and dystroglycan systems and provides new insight into the molecular mechanisms by which TCAP-1 regulates cytoskeletal dynamics in hippocampal neurons. The TCAP-dystroglycan system may represent a novel mechanism associated with the regulation of hippocampal-function.


Assuntos
Proteínas Contráteis/metabolismo , Citoesqueleto/metabolismo , Distroglicanas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células Piramidais/metabolismo , Estatmina/metabolismo , Tenascina/metabolismo , Animais , Western Blotting , Filaminas , Imunofluorescência , Hipocampo/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Neurogênese/fisiologia
5.
Int J Obes (Lond) ; 35(2): 198-207, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20548307

RESUMO

OBJECTIVE: Estrogen (E(2)) has an inhibitory effect on food intake by acting centrally in the hypothalamus, although it is not clear which hypothalamic neurons are involved in this process. Earlier studies from our lab and others have implicated neuropeptide Y (NPY) as an important central anorexigenic target of E(2). This study was designed to investigate whether E(2) can directly regulate NPY secretion and examine the cellular mechanisms and receptors responsible for this anorexigenic action of E(2). DESIGN: Clonal, murine, hypothalamic neuronal cell models, mHypoE-42 and mHypoA-2/12, were investigated for NPY secretory responses to 17ß-estradiol (E(2)) in the presence or absence of pharmacological inhibitors directed against the phosphatidylinositol-3-kinase (PI3K), mitogen-activated protein kinase (MAPK) and AMP-activated kinase (AMPK) pathways or to estrogen receptor (ER) specific agonists/antagonists. MEASUREMENTS: The presence of hypothalamic markers and characterization of neuronal cell lines was completed with polymerase chain reaction. NPY levels were measured using an enzyme immunoassay (EIA). The expression of ER-α and caveolin-1 was analyzed using immunocytochemistry. RESULTS: E(2) significantly decreased NPY secretion in both the mHypoE-42 and mHypoA-2/12 neurons. The E(2)-mediated repression of NPY secretion in the mHypoE-42 and mHypoA-2/12 neurons required ER-α, but not ER-ß, as shown by studies using an ER-specific agonist/antagonists. Additionally, using immunocytochemistry we detected colocalization of ER-α and the membrane-associated signaling protein caveolin-1. Importantly, using E(2)-conjugated bovine serum albumin (E(2)-BSA) and ER antagonists, we were able to show that the E(2)-mediated decrease in NPY secretion occurred through membrane-bound ER-α. Finally, using a combination of pharmacological inhibitors, we found that inhibition of the PI3K or AMPK pathway blocked the E(2)-mediated decrease in NPY secretion. CONCLUSION: These findings indicate that the central anorexigenic action of E(2) occurs at least partially through hypothalamic NPY-synthesizing neurons. This regulation of NPY secretion occurs through rapid signaling mechanisms through membrane bound ER-α.


Assuntos
Regulação do Apetite/fisiologia , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Hipotálamo/efeitos dos fármacos , Neuropeptídeo Y/metabolismo , Transdução de Sinais/fisiologia , Animais , Regulação do Apetite/genética , Biomarcadores , Bovinos , Caveolina 1/metabolismo , Linhagem Celular , Receptor alfa de Estrogênio/genética , Hipotálamo/metabolismo , Imuno-Histoquímica , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeo Y/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética
6.
Neuroscience ; 170(1): 54-66, 2010 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-20619320

RESUMO

The hippocampus is a multifaceted, complex brain structure considered to be the learning center. The use of primary hippocampal cell cultures has uncovered important cellular mechanisms involved in overall physiological function. Yet, the use of primary culture is inherently difficult, and the lack of immortalized cell lines from the murine hippocampus for mechanistic studies at the molecular level is evident. We have immortalized cell lines from embryonic (E18) and adult-derived hippocampal primary cell culture using retroviral infection of SV40 T-antigen. Four clonal embryonic lines, mHippoE-2, mHippoE-5, mHippoE-14, mHippoE-18, and one mixed adult line, mHippoA-mix, exhibited neuronal morphologies with neurite extensions and expression of neuronal markers, with unique gene expression profiles. We used these cell models to study the neuroprotective effects of 17beta-estradiol (E2) on glutamate-induced neurotoxicity. The cell lines express a relevant array of genes and receptors suggested to play a role in neuroprotection, including estrogen receptors ERalpha, ERbeta, and GPR30. We find that pretreatment with E2 (10 or 100 nM) for 24 h significantly reduced cell death induced by glutamate mHippoE-14 and mHippoE-18 cells, but not the mHippoA-mix. Using 24 h pretreatment with the specific estrogen receptor (ER) agonists, 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT) and diarylpropionitrile, 2,3-bis(4-Hydroxyphenyl)-propionitrile (DPN), we linked the E2-mediated neuroprotection to ERalpha, but only in the mHippoE-18 cells. Since E2 activated both PI3K/Akt and STAT3 signaling pathways, we also tested whether the membrane-bound E2 receptor GPR30 was involved in its neuroprotective action. Pretreatment with the GPR30 agonist G-1 (10 and 100 nM) for 1 h, but not 24 h, significantly attenuated cell death in both mHippoE-14 and mHippoE-18 cells. The use of specific ER antagonist ICI 182780 and GPR30 antagonist G-15 linked these effects to both ER and GPR30 receptors. This is the first evidence that GPR30 may play a role in the protective effects of estrogen in hippocampal neurons.


Assuntos
Estradiol/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/fisiologia , Hipocampo/fisiologia , Fármacos Neuroprotetores/farmacologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/metabolismo , Gravidez , Receptores de Estrogênio
7.
Neuroscience ; 162(4): 1134-40, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19463905

RESUMO

Significant information on reproductive function has been generated based on the rat model, including many seminal discoveries. Yet little is known about the molecular and cellular events involved in control of reproductive function, mainly due to the pervasive lack of cell models from rat. We have therefore generated a wide array of cell lines using primary cell culture from the rat hypothalamus. Immortalization of the primary cells was achieved through retroviral transfer of T-antigen, followed by selection with geneticin. The mixed cell populations were subcloned and each clonal cell line was analyzed for expression of specific cellular markers. Each line has a distinct phenotypic profile, with expression of key neuroendocrine markers. We have functionally analyzed two clonal cell lines, rHypoE-7 and rHypoE-8, for hormones implicated in the control of gonadotropin-releasing hormone neuronal function through melatonin, specifically kisspeptin (KISS) and RF-amide-related peptide-3 (RFRP-3, the mammalian ortholog of the avian gonadotropin-inhibiting hormone, GnIH). We detected functional melatonin receptor activity, as each cell line exhibited inhibition of forskolin-stimulated 3'-5'-cyclic adenosine monophosphate (cAMP) accumulation. Upon treatment with 10 nM melatonin, we found that KISS gene expression was decreased in the rHypoE-8 cell line, while RFRP-3 was increased in the rHypoE-7 cell line. These results are in accordance with the differential regulatory functions of these two peptides, particularly on GnRH neuronal control. These cell lines will serve as novel tools for the analysis of the cellular and molecular mechanisms involved in hypothalamic control of a number of physiological processes described in the rat animal model.


Assuntos
Linhagem Celular , Células Clonais , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hipotálamo/citologia , Melatonina/fisiologia , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Proteínas/metabolismo , Animais , Embrião de Mamíferos/citologia , Kisspeptinas , Melatonina/farmacologia , Neurônios/citologia , Neuropeptídeos/genética , RNA Mensageiro/metabolismo , Ratos , Receptores de Melatonina/metabolismo
8.
Neuroendocrinology ; 89(3): 267-75, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18957841

RESUMO

The gut peptide ghrelin is expressed within neurons of the hypothalamus. Using a hypothalamic cell line, mHypoE-38 neurons, the effect of insulin on preproghrelin gene expression was assayed. These cells contain neuron-specific markers, preproghrelin and the insulin receptor. We determined that insulin has direct effects on preproghrelin gene expression. Insulin (10 nM) stimulated protein kinase B (Akt) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation from 5 to 60 min and 5 min, respectively, and led to repression of preproghrelin gene expression at 2 h. Pharmacological inhibitors to phosphoinositide-3-kinase (PI3-K; LY294002) and MEK (PD98059) demonstrated that basal ghrelin gene expression is regulated by the PI3-K pathway and requires the mitogen-activated protein kinase pathway for insulin-stimulated preproghrelin repression. These results demonstrate that insulin has a direct effect on hypothalamic neurons to decrease preproghrelin gene expression through classic insulin pathways.


Assuntos
Regulação para Baixo , Grelina/genética , Hipotálamo/metabolismo , Insulina/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/agonistas , Animais , Linhagem Celular , Cromonas/farmacologia , Flavonoides/farmacologia , Hipotálamo/efeitos dos fármacos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Camundongos , Morfolinas/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Receptor de Insulina/metabolismo
9.
J Med Genet ; 41(5): 350-3, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15121772

RESUMO

The agouti related protein (AgRP) exerts its anabolic effects on food intake by antagonising the alpha-melanocyte stimulating hormone (alpha-MSH) at its receptors, melanocortin receptors 3 and 4 (MC3R and MC4R). A single nucleotide polymorphism (SNP) in the promoter of the human AgRP (hAgRP), -38C>T, was associated with low body fatness. The -38T allele that was associated with low body fatness also resulted in lower promoter activity. Here we report a novel SNP, -3019G>A, again in the promoter of hAgRP, which is in complete linkage disequilibrium (LD) with the -38C>T SNP (linked alleles: -3019A/-38T and -3019G/-38C). Functional analyses in a human adrenal and two mouse hypothalamus cell lines showed that the -3019A allele had significantly higher promoter activity. Hence, the two linked alleles (-3019A and -38T) had opposite effects on promoter function and yet they were both associated with low body fatness. The region encompassing the -38C>T SNP had approximately 1000-fold higher activity than the region encompassing the -3019G>A SNP, potentially determining the net functional effect between these two SNPs.


Assuntos
Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Proteínas/genética , Proteína Relacionada com Agouti , Animais , Linhagem Celular , Feminino , Frequência do Gene , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Desequilíbrio de Ligação , Masculino , Camundongos , Ativação Transcricional
10.
Endocrinology ; 142(11): 4711-20, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11606436

RESUMO

The pineal hormone melatonin plays an important role in the neuroendocrine control of reproductive physiology, but its effects on hypothalamic GnRH neurons are not yet known. We have found that GT1-7 GnRH-secreting neurons express membrane-bound G protein-coupled melatonin receptors, mt1 (Mel-1a) and MT2 (Mel-1b) as well as the orphan nuclear receptors ROR alpha and RZR beta. Melatonin (1 nM) significantly downregulates GnRH mRNA levels in a 24-h cyclical manner, an effect that is specifically inhibited by the melatonin receptor antagonist luzindole (10 microM). Repression of GnRH gene expression by melatonin appears to occur at the transcriptional level and can be mapped to the GnRH neuron-specific enhancer located within the 5' regulatory region of the GnRH gene. Using transient transfection of GT1-7 cells, downregulation of GnRH gene expression by melatonin was further localized to five specific regions within the GnRH enhancer including -1827/-1819, -1780/-1772, -1746/-1738, -1736/-1728, and -1697/-1689. Interestingly, the region located at -1736/-1728 includes sequences that correspond to two direct repeats of hexameric consensus binding sites for members of the ROR/RZR orphan nuclear receptor family. To begin to dissect the mechanisms involved in the 24-h cyclical regulation of GnRH transcription, we have found that melatonin (10 nM) induces rapid internalization of membrane-bound mt1 receptors through a beta-arrestin 1-mediated mechanism. These results provide the first evidence that melatonin may mediate its neuroendocrine control on reproductive physiology through direct actions on the GnRH neurons of the hypothalamus, both at the level of GnRH gene expression and through the regulation of G protein-coupled melatonin receptors.


Assuntos
Ritmo Circadiano , Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Melatonina/fisiologia , Neurônios/metabolismo , Animais , Sequência de Bases/genética , Linhagem Celular , Elementos Facilitadores Genéticos/genética , Elementos Facilitadores Genéticos/fisiologia , Melatonina/farmacologia , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Melatonina , Transcrição Gênica/fisiologia , Triptaminas/farmacologia
11.
Mol Endocrinol ; 14(2): 212-28, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10674395

RESUMO

The physiological actions of nitric oxide (NO) as a signaling molecule in endothelial and brain cells and as a toxic molecule used by activated immune cells have been the focus of a wide range of studies. Nevertheless, the downstream effector molecules of this important neuromodulator are not well understood. We have previously demonstrated that expression of the gene for the reproductive neuropeptide, GnRH, is repressed by the glutamate/NO/cyclic GMP (cGMP) signal transduction pathway through cGMP-dependent protein kinase in the hypothalamic GnRH-secreting neuronal cell line GT1-7. This repression localized within a previously characterized 300-bp neuron-specific enhancer. Here, we find that mutation of either of two adjacent elements within the enhancer eliminates repression by this pathway. An AT-rich sequence located at -1695 has homology to the octamer motif known to bind POU-homeodomain proteins, while the adjacent element at -1676 has homology to the C/EBP (CCAAT/enhancer-binding protein) protein family consensus sequence. Antibody supershift assays reveal that one of the proteins bound at the -1695 sequence is Oct-1, and one of the proteins bound to the element at -1676 is C/EBPbeta. These two proteins can bind simultaneously to the adjacent -1695 and -1676 binding sites in vitro. In nuclear extracts of GT1-7 cells treated with an NO donor, the intensity of the Oct-1 complex is increased. However, although Western blot analysis indicates that neither Oct-1 nor C/EBPbeta protein levels are increased, the relative binding affinity of Oct-1 is increased. Dephosphorylation of the nuclear extracts decreases binding of the Oct-1 complex to the -1695 site only in NO donor-treated extracts. Thus, we conclude that Oct-1 and C/EBPbeta are both downstream transcriptional regulators involved in the repression of GnRH gene expression by the glutamate/NO/ cGMP signal transduction pathway.


Assuntos
GMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Hormônio Liberador de Gonadotropina/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Proteínas Estimuladoras de Ligação a CCAAT , Linhagem Celular/efeitos dos fármacos , GMP Cíclico/farmacologia , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Ácido Glutâmico/metabolismo , Hormônio Liberador de Gonadotropina/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/metabolismo , Fator C1 de Célula Hospedeira , Hipotálamo/citologia , Camundongos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Nitroprussiato/farmacologia , Fator 1 de Transcrição de Octâmero , Ratos , Transdução de Sinais
12.
Endocrinology ; 140(11): 5045-53, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10537130

RESUMO

Estrogen has wide-ranging and complex effects on the reproductive axis, which are often difficult to interpret from in vivo studies. Estrogen negatively regulates tonic GnRH synthesis and also plays a pivotal role in the positive regulation of GnRH necessary for the preovulatory surge. To dissect the mechanisms by which these divergent effects occur, we attempted to observe the direct action of estrogen on the regulation of GnRH messenger RNA (mRNA) levels using the well characterized, GnRH-secreting, hypothalamic cell line, GT1-7. Using RT-PCR, we first investigated estrogen receptor transcript expression in GT1-7 neurons. We found that the GT1-7 cells express both estrogen receptor-alpha (ERalpha) and the recently described ERbeta mRNAs. We also detected the presence of both receptor subtypes in the GT1-7 neurons by Western blot analysis using specific ER antibodies. By Northern blot analysis of total GT1-7 RNA, we found that 17beta-estradiol (1 nM) down-regulates GnRH mRNA levels to approximately 55% of basal levels over a 48-h time course. This effect appears to occur specifically through an ER-mediated mechanism, as ICI 182,780, a complete ER antagonist, blocks the repression of GnRH mRNA levels by estradiol. The recently reported ERalpha-specific agonist/ERbeta-specific antagonist 2,2-bis-(p-hydroxyphenyl-1,1,1-trichloroethane (HPTE), a methoxychlor metabolite, also down-regulated GnRH gene expression. The repression of GnRH mRNA levels appears to occur at the transcriptional level, as simian virus 40 T antigen mRNA expression, which is under the control of 2.3 kb of the rat GnRH 5'-regulatory region, mimics the down-regulation of GnRH after treatment with estradiol. As the rat GnRH regulatory region in GT1-7 neurons does not appear to harbor a classic estrogen response element, the mechanism involved in the repression of GnRH has yet to be determined. These results suggest that estradiol directly regulates GnRH gene expression at the level of the GnRH neuron and may exert its neuroendocrine control through direct interaction with specific receptors expressed in these cells.


Assuntos
Estradiol/farmacologia , Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Neurônios/metabolismo , Receptores de Estrogênio/genética , Animais , Antígenos Transformantes de Poliomavirus/genética , Northern Blotting , Linhagem Celular , Sondas de DNA , Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Fulvestranto , Hormônio Liberador de Gonadotropina/metabolismo , Camundongos , Fenóis/farmacologia , RNA Mensageiro/metabolismo , Ratos , Receptores de Estrogênio/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica/efeitos dos fármacos
13.
Endocrinology ; 139(3): 1108-14, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9492044

RESUMO

Hypothalamic GnRH secretory neurons are precisely regulated by circulating gonadal steroids. However, the question of whether these cells are directly responsive to steroid hormones remains a central and controversial issue in reproductive science. In the present study, we demonstrate the expression of androgen receptor (AR) in a mouse hypothalamic GnRH-secreting cell line, GT1-7. AR messenger RNA was detected by Northern blot analysis of 10 microg total cellular RNA. Western blot analysis revealed a 110K AR immunoreactive band, and saturation binding analysis confirmed the presence of a high affinity low capacity androgen binding entity (Kd = 0.06 nM; Bmax = 12.4 fmol/mg protein). In addition, GT1-7 cells were found to express ARA70, an AR-specific coactivator that has been reported to enhance transactivational activity of the AR. GT1-7 cells transiently transfected with an androgen responsive MMTV-luciferase reporter construct displayed a 4.2-fold induction of luciferase reporter gene activity by 1 nM 5alpha-dihydrotestosterone (DHT), further demonstrating the presence of a functional AR. Treatment of GT1-7 cells with 1 or 10 nM DHT resulted in approximately 55% reduction in GnRH messenger RNA measured at 24 and 36 h after treatment. This repression was completely blocked by hydroxyflutamide, an AR antagonist. These results provide the first demonstration that androgen acts directly through an AR-mediated pathway to repress GnRH gene expression in hypothalamic GnRH-secreting neurons.


Assuntos
Di-Hidrotestosterona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Neurônios/metabolismo , Animais , Células Cultivadas , Flutamida/análogos & derivados , Flutamida/farmacologia , Humanos , Camundongos , RNA Mensageiro/análise , Receptores Androgênicos/análise
14.
EMBO J ; 15(3): 538-47, 1996 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8599937

RESUMO

The key roles of the excitatory neurotransmitter glutamate and its second messengers, nitric oxide (NO) and cGMP, in long-term potentiation and neural plasticity are well documented. However, complex functions such as memory are likely to require long term changes in synaptic efficacy which require gene expression and protein synthesis. Here we demonstrate that the glutamate receptor agonist, N-methyl-D-aspartic acid (NMDA), nitric oxide (NO) and cGMP each repress expression of the gonadotropin-releasing hormone (GnRH) gene in the hypothalamic cell line, GT1. This repression is dependent upon signals from NMDA receptors activating NO synthase to synthesize NO. In turn NO induces guanylyl cyclase to synthesize cGMP, activating cGMP- dependent protein kinase. Repression requires elevation of calcium because it only occurs in the presence of calcium ionophore or with release of intracellular calcium. Repression also requires protein synthesis. Activation of this pathway specifically represses expression of a reporter gene containing the regulatory region of the GnRH gene in transfected GT1 cells, indicating that repression occurs at the transcriptional level. Furthermore the target for transcriptional repression is a 300 bp neuron-specific enhancer found 1.5 kb upstream of the GnRH gene which is sufficient to confer repression to a heterologous promoter. Thus the NMDA/NO/cGMP neurotransmitter signal transduction pathway controls not only synaptic function but also neuron-specific gene expression.


Assuntos
GMP Cíclico/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , N-Metilaspartato/farmacologia , Óxido Nítrico/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Animais , Sequência de Bases , Cálcio/metabolismo , Linhagem Celular , GMP Cíclico/farmacologia , Primers do DNA/genética , Agonistas de Aminoácidos Excitatórios/farmacologia , Expressão Gênica/efeitos dos fármacos , Guanilato Ciclase/metabolismo , Camundongos , Dados de Sequência Molecular , Óxido Nítrico Sintase/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Transfecção
15.
Mol Endocrinol ; 9(4): 467-77, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7659090

RESUMO

The molecular mechanisms specifying gene expression in individual neurons of the mammalian central nervous system have been difficult to study due to the cellular complexity of the brain and the absence of cultured model systems representing differentiated central nervous system neurons. We have developed clonal, differentiated, neuronal tumor cell lines of the hypothalamic GnRH-producing neurons by targeting tumorigenesis in transgenic mice. These cells (GT1 cells) provide a model system for molecular studies of GnRH gene regulation. Here we present the identification and characterization of a neuron-specific enhancer responsible for directing expression of the rat GnRH gene in GT1 hypothalamic neurons. This approximately 300 base pair (bp) upstream region (-1571 to -1863) confers enhancer activity to a short -173-bp GnRH promoter or to a heterologous promoter only in GT1 cells. The enhancer is bound by multiple GT1 nuclear proteins over its entire length. Deletion of more than 30 bp from either end dramatically reduces activity, and even large internal fragments carrying seven of the eight DNAse I-protected elements show decreased activity. Scanning replacement mutations demonstrate that several of the internal elements are required for activity of the enhancer. Thus, the GnRH gene is targeted to hypothalamic neurons by a complex multicomponent enhancer that relies on the interaction of multiple nuclear-protein binding enhancer elements.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Neurônios/metabolismo , Animais , Antígenos Transformantes de Poliomavirus/fisiologia , Sequência de Bases , Transformação Celular Viral , Neoplasias Hipotalâmicas/genética , Neoplasias Hipotalâmicas/patologia , Neoplasias Hipotalâmicas/virologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutagênese , Especificidade de Órgãos , Regiões Promotoras Genéticas , Ratos , Proteínas Recombinantes de Fusão/biossíntese , Vírus 40 dos Símios/genética , Células Tumorais Cultivadas
16.
Hum Mutat ; 5(1): 28-33, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7537149

RESUMO

A large Manitoba Hutterite kindred with X-linked receptor negative complete androgen insensitivity syndrome (CAIS) was studied. In attempts to identify all carriers of the syndrome in this kindred, using the androgen receptor (AR) cDNA, we have found a novel diagnostic MspI polymorphic pattern, which cosegregates with the disease. This polymorphism was not detected in 79 unrelated X-chromosomes of which 22 were from Hutterite controls. We were able to localize the polymorphism to exon 4, which is known to encode part of the androgen receptor hormone binding domain. A single base substitution (T-->C) was detected, which creates a new MspI site. This novel transition mutation replaces Leu-676 with Pro at a site which is conserved in numerous members of the steroid receptor gene family. Sequencing all 8 exons of the AR revealed the Leu-676-->Pro mutation as the only change in the primary structure of the receptor. Transfection of COS-1 cells with an expression vector of the mutant AR demonstrates that this point mutation of nucleotide 2558 abolishes receptor binding activity. The mutation can easily be detected by MspI digestion of the polymerase chain reaction (PCR) amplified exon 4 product.


Assuntos
Síndrome de Resistência a Andrógenos/genética , Mutação Puntual , Receptores Androgênicos/genética , Sequência de Aminoácidos , Síndrome de Resistência a Andrógenos/metabolismo , Animais , Sequência de Bases , Células Cultivadas , Pré-Escolar , DNA Complementar/genética , Desoxirribonuclease HpaII , Desoxirribonucleases de Sítio Específico do Tipo II , Etnicidade/genética , Feminino , Triagem de Portadores Genéticos , Humanos , Masculino , Manitoba , Dados de Sequência Molecular , Linhagem , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Receptores Androgênicos/metabolismo , Transfecção , Cromossomo X
17.
Fertil Steril ; 60(2): 366-8, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8339840

RESUMO

Nonaromatizable androgens, administered in high doses to an adult patient with partial androgen insensitivity, failed to result in a change in phallic size despite a clear decline in SHBG and gonadotropin levels. These findings raise the question of differential tissue sensitivity to androgens. Because ancillary laboratory testing does not predict reliably the genital response, a therapeutic trial should be advocated in such cases.


Assuntos
Androgênios/farmacologia , Adulto , Ligação Competitiva , Resistência a Medicamentos , Fibroblastos/metabolismo , Genitália Masculina/metabolismo , Genitália Masculina/patologia , Gonadotropinas/sangue , Humanos , Masculino , Pênis/anormalidades , Receptores Androgênicos/metabolismo , Valores de Referência , Globulina de Ligação a Hormônio Sexual/análise , Pele/metabolismo , Pele/patologia
18.
J Neurol Sci ; 112(1-2): 133-8, 1992 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1469423

RESUMO

Herein we describe a family with X-linked spinal and bulbar muscular atrophy (SBMA or Kennedy's disease), an adult onset neuromuscular disease characterized by slow progression, predominant proximal and bulbar muscle weakness. One frequent association is the appearance of gynecomastia. This disorder was previously shown to be linked to the locus DXYS1 on the proximal long arm of the X chromosome. Recently, a report implicated a mutation at the N-terminus of the androgen receptor gene involving amplification of CAG repeats as the cause of X-linked SBMA. We studied this region of the androgen receptor in a kindred clinically suspected but not confirmed of having X-linked SBMA by the polymerase chain reaction (PCR) followed by Southern analysis and DNA sequencing. The mutated allele was found to have an increased number of 51 CAG repeats confirming the clinical diagnosis of SBMA. Normal individuals revealed 23 repeat numbers within the normal range, while another unrelated X-linked SBMA patient had an enlarged CAG repeat region. The carrier or disease status could be established or confirmed in 12 individuals of this family on the basis of detecting normal and disease alleles reflected by the number of CAG repeats.


Assuntos
Ligação Genética , Atrofia Muscular Espinal/genética , Receptores Androgênicos/genética , Sequências Repetitivas de Ácido Nucleico/genética , Cromossomo X , Adulto , Sequência de Aminoácidos , Sequência de Bases , Southern Blotting , Eletroforese em Gel de Poliacrilamida , Heterozigoto , Humanos , Masculino , Dados de Sequência Molecular , Linhagem , Reação em Cadeia da Polimerase
19.
Am J Hum Genet ; 51(1): 143-55, 1992 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1609793

RESUMO

We have discovered two different point mutations in a single codon of the X-linked androgen-receptor (AR) gene in two pairs of unrelated families who have complete androgen insensitivity (resistance) associated with different AR phenotypes in their genital skin fibroblasts. One mutation is a C-to-T transition at a CpG sequence near the 5' terminus of exon 6; it changes the sense of codon 773 from arginine to cysteine, ablates specific androgen-binding activity at 37 degrees C, and eliminates a unique KpnI site at the intron-exon boundary. The other mutation is a G-to-A transition that changes amino acid 773 to histidine and eliminates an SphI site. This mutant AR has a normal androgen-binding capacity at 37 degrees C but has a reduced affinity for androgens and is thermolabile in their presence. Transient transfection of COS cells with cDNA expression vectors yielded little androgen-binding activity at 37 degrees C from Arg773Cys and abundant activity with abnormal properties from Arg773His, thereby providing the pathogenicity of both sequence alterations. This conclusion coincides with the following facts about evolutionary preservation of the position homologous to Arg773 in the AR: it is occupied by Arg or lysine in the progesterone, glucocorticoid, and mineralocorticoid receptors, and it is within a 14-amino-acid region of their steroid-binding domains that share approximately 85% amino acid identity.


Assuntos
Transtornos do Desenvolvimento Sexual/genética , Receptores Androgênicos/química , Adolescente , Arginina , Sequência de Bases , Canadá , Cisteína , Desoxirribonucleases de Sítio Específico do Tipo II , Transtornos do Desenvolvimento Sexual/metabolismo , Feminino , Histidina , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutação , Linhagem , Fenótipo , Reação em Cadeia da Polimerase , Receptores Androgênicos/genética , Ativação Transcricional
20.
Am J Med Genet ; 40(4): 493-9, 1991 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-1720929

RESUMO

We have discovered in the X-linked androgen receptor gene a single nucleotide substitution that is the putative cause of complete androgen insensitivity (resistance) in a family with affected individuals in 2 generations. Earlier studies on the family indicated co-segregation of mutant phenotype and the RFLPs at the loci DXS1 and DXYS1. The mutation is an adenine-to-thymine transversion in exon 8 that changes the sense of codon 882 from lysine to an amber (UAG) translation termination signal. The substitution creates a recognition sequence for the restriction endonuclease MaeI: this permits ready recognition of hemizygotes and heterozygotes after amplification of genomic exon 8 by the polymerase chain reaction. The mutation predicts the synthesis of a truncated receptor that lacks 36 amino acids at the carboxy terminus of its 252-amino acid androgen-binding domain. The cultured genital skin fibroblasts of the one affected patient examined have normal levels of androgen receptor mRNA, but negligible androgen-receptor binding activity. These results accord with a variety of data from spontaneous and artificial mutations indicating that all portions of the steroid binding domain contribute to normal steroid binding by a steroid receptor.


Assuntos
Androgênios/metabolismo , Mapeamento Cromossômico , Biossíntese de Proteínas/genética , Receptores Androgênicos/genética , Sequência de Aminoácidos , Sequência de Bases , Criança , DNA/isolamento & purificação , Éxons/fisiologia , Feminino , Ligação Genética , Humanos , Immunoblotting , Dados de Sequência Molecular , Mutação , Linhagem , Reação em Cadeia da Polimerase , RNA/isolamento & purificação , Cromossomo X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...