Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Mol Endocrinol ; 64(3): 165-179, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31990658

RESUMO

Many sex differences in liver gene expression originate in the brain, depend on GH secretion and may underlie sex disparities in hepatic disease. Because epigenetic mechanisms may contribute, we studied promoter methylation and microRNA abundance in the liver, associated with expression of sexual dimorphic genes in mice with selective disruption of the dopamine D2 receptor in neurons (neuroDrd2KO), which decreases hypothalamic Ghrh, pituitary GH, and serum IGFI and in neonatally androgenized female mice which have increased pituitary GH content and serum IGFI. We evaluated mRNA levels of the female predominant genes prolactin receptor (Prlr), alcohol dehydrogenase 1 (Adh1), Cyp2a4, and hepatocyte nuclear transcription factor 6 (Hnf6) and the male predominant gene, Cyp7b1. Female predominant genes had higher mRNA levels compared to males, but lower methylation was only detected in the Prlr and Cyp2a4 female promoters. In neuroDrd2KO mice, sexual dimorphism was lost for all genes; the upregulation (feminization) of Prlr and Cyp2a4 in males correlated with decreased methylation of their promoters, and the downregulation (masculinization) of Hnf-6 mRNA in females correlated inversely with its promoter methylation. Neonatal androgenization of females evoked a loss of sexual dimorphism only for the female predominant Hnf6 and Adh1 genes, but no differences in promoter methylation were found. Finally, mmu-miR-155-5p, predicted to target Cyp7b1 expression, was lower in males in association with higher Cyp7b1 mRNA levels compared to females and was not modified in neuroDrd2KO or TP mice. Our results suggest specific regulation of gene sexually dimorphic expression in the liver by methylation or miRNAs.


Assuntos
Álcool Desidrogenase/genética , Hidrocarboneto de Aril Hidroxilases/genética , Família 2 do Citocromo P450/genética , Família 7 do Citocromo P450/genética , Hormônio do Crescimento/farmacologia , Fator 6 Nuclear de Hepatócito/genética , Receptores da Prolactina/genética , Esteroide Hidroxilases/genética , Álcool Desidrogenase/metabolismo , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Família 2 do Citocromo P450/metabolismo , Família 7 do Citocromo P450/metabolismo , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/fisiologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio do Crescimento/metabolismo , Fator 6 Nuclear de Hepatócito/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Receptores da Prolactina/metabolismo , Caracteres Sexuais , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esteroide Hidroxilases/metabolismo
2.
Cell Mol Neurobiol ; 39(2): 169-180, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30656469

RESUMO

A multistep signaling cascade originates in brain centers that regulate hypothalamic growth hormone-releasing hormone (Ghrh) and somatostatin expression levels and release to control the pattern of GH secretion. This process is sexually fine-tuned, and relays important information to the liver where GH receptors can be found. The temporal pattern of pituitary GH secretion, which is sex-specific in many species (episodic in males and more stable in females), represents a major component in establishing and maintaining the sexual dimorphism of hepatic gene transcription. The liver is sexually dimorphic exhibiting major differences in the profile of more than 1000 liver genes related to steroid, lipid, and foreign compound metabolism. Approximately, 90% of these sex-specific liver genes were shown to be primarily dependent on sexually dimorphic GH secretory patterns. This proposes an interesting scenario in which the central nervous system, indirectly setting GH profiles through GHRH and somatostatin control, regulates sexual dimorphism of liver activity in accordance with the need for sex-specific steroid metabolism and performance. We describe the influence of the loss of sexual dimorphism in liver gene expression due to altered brain function. Among other many factors, abnormal brain sexual differentiation, xenoestrogen exposure and D2R ablation from neurons dysregulate the GHRH-GH axis, and ultimately modify the liver capacity for adaptive mechanisms. We, therefore, propose that an inefficient brain control of the endocrine growth axis may underlie alterations in several metabolic processes through an indirect influence of sexual dimorphism of liver genes.


Assuntos
Encéfalo/fisiopatologia , Sistema Endócrino/fisiopatologia , Hepatopatias/fisiopatologia , Fígado/fisiopatologia , Caracteres Sexuais , Animais , Epigênese Genética , Feminino , Humanos , Hepatopatias/genética , Masculino
3.
Endocrinology ; 156(3): 1040-51, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25545383

RESUMO

Liver sexual gene dimorphism, which depends mainly on specific patterns of GH secretion, may underlie differential susceptibility to some liver diseases. Because GH and prolactin secretion are regulated by dopaminergic pathways, we studied the participation of brain and lactotrope dopamine 2 receptors (D2Rs) on liver gene sexual dimorphism, to explore a link between the brain and liver gene expression. We used global D2R knockout mice (Drd2(-/-)) and conducted a functional dissection strategy based on cell-specific Drd2 inactivation in neurons (neuroDrd2KO) or pituitary lactotropes. Disruption of neuronal D2Rs (which impaired the GH axis) decreased most of male or female-predominant class I liver genes and increased female-predominant class II genes in males, consistent with the positive (class I) or negative (class II) regulation of these genes by GH. Notably, sexual dimorphism was lost for class I and II genes in neuroDrd2KO mice. Disruption of lactotrope D2Rs did not modify class I or II genes in either sex, because GH axis was preserved. But surprisingly, 1 class II gene (Prlr) and female-predominant class I genes were markedly up-regulated in lacDrd2KO females, pointing to direct or indirect effects of prolactin in the regulation of selected female-predominant liver genes. This suggestion was strengthened in the hyperprolactinemic Drd2(-/-) female mouse, in which increased expression of the same 4 liver genes was observed, despite a decreased GH axis. We hereby demonstrate endocrine-mediated D2R actions on sexual dimorphic liver gene expression, which may be relevant during chronic dopaminergic medications in psychiatric disease.


Assuntos
Encéfalo/metabolismo , Fígado/metabolismo , Hipófise/metabolismo , Receptores de Dopamina D2/metabolismo , Animais , Feminino , Regulação da Expressão Gênica/fisiologia , Hormônio do Crescimento/sangue , Hormônio do Crescimento/metabolismo , Lactotrofos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Proteínas/genética , Proteínas/metabolismo , Receptores de Dopamina D2/genética , Caracteres Sexuais
4.
Endocrinology ; 155(3): 829-39, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24424036

RESUMO

Prolactin, a pleiotropic hormone secreted by lactotropes, has reproductive and metabolic functions. Chronically elevated prolactin levels increase food intake, but in some hyperprolactinemic states such as in the global dopamine D2 receptor (D2R) knockout mouse, food intake is not increased. Here, we conduct a cell-specific genetic dissection study using conditional mutant mice that selectively lack D2Rs from pituitary lactotropes (lacDrd2KO) to evaluate the role of elevated prolactin levels without any confounding effect of central D2Rs on motor and reward mechanisms related to food intake. LacDrd2KO female mice exhibited chronic hyperprolactinemia, pituitary hyperplasia, and a preserved GH axis. In addition, lacDrd2KO female but not male mice showed increased food intake by 3 months of age, and from 5 months onward their body weights were heavier. Marked increments in fat depots, adipocyte size, serum triglycerides, and nonesterified fatty acid levels and a decrease in lipolytic enzymes in adipose tissue were seen. Furthermore, lacDrd2KO female mice had glucose intolerance but a preserved response to insulin. In the hypothalamus, Npy mRNA expression was increased, and Pomc and Ppo mRNA levels were unaltered (in contrast to results in global D2R knockout mice). Thus, the orexigenic effect of prolactin and its action on hypothalamic Npy expression were fully evidenced, leading to increased food intake and adiposity. Our results highlight the metabolic role of prolactin and illustrate the value of studying cell-specific mutant mice to disentangle the pathophysiological mechanisms otherwise masked in null allele mutants or in animals treated with pervasive pharmacological agents.


Assuntos
Adiposidade/genética , Peso Corporal/genética , Ingestão de Alimentos/genética , Hipófise/metabolismo , Receptores de Dopamina D2/genética , Tecido Adiposo/metabolismo , Animais , Ciclo Estral , Feminino , Genótipo , Intolerância à Glucose , Teste de Tolerância a Glucose , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prolactina/sangue
5.
Mol Cell Endocrinol ; 382(2): 825-34, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24239981

RESUMO

Neonatal androgenization masculinizes the GH axis and thus may impact on liver gene regulation. Neonatal testosterone administration to female mice decreased (defeminized) female predominant GH-dependent liver gene expression (Hnf6, Adh1, Prlr, Cyp3a41) and did not modify male predominant genes (Cyp7b1, Cyp4a12, Slp). Female predominance of Cis mRNA, an inhibitor of episodic GH signaling pathway, was unaltered. At birth, Cyp7b1 promoter exhibited a higher methylation status in female livers, while the Hnf6 promoter was equally methylated in both sexes; no differences in gene expression were detected at this age. In adulthood, consistent with sex specific predominance, lower methylation status was determined for the Cyp7b1 promoter in males, and for the Hnf6 promoter in females, and this last difference was prevented by neonatal androgenization. Therefore, early steroid treatment or eventually endocrine disruptor exposure may alter methylation status and sexual dimorphic expression of liver genes, and consequently modify liver physiology in females.


Assuntos
Androgênios/farmacologia , Hormônio do Crescimento/genética , Fator 6 Nuclear de Hepatócito/genética , Fígado/efeitos dos fármacos , Esteroide Hidroxilases/genética , Testosterona/farmacologia , Animais , Animais Recém-Nascidos , Família 7 do Citocromo P450 , Metilação de DNA/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hormônio do Crescimento/metabolismo , Fator 6 Nuclear de Hepatócito/metabolismo , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Masculino , Camundongos , Regiões Promotoras Genéticas/efeitos dos fármacos , Caracteres Sexuais , Transdução de Sinais , Esteroide Hidroxilases/metabolismo , Virilismo/genética
6.
Toxicol Lett ; 213(3): 325-31, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22842222

RESUMO

The hypothalamic-growth hormone (GH)-liver axis represents a new concept in endocrine regulation of drug toxicity. Preponderant sex differences are found in liver gene expression, mostly dependent on the sexually dimorphic pattern of GH secretion which is set during the neonatal period by gonadal steroids. We tested if GH-dependent sexually dimorphic liver enzymes and proteins was perturbed by neonatal Bisphenol A (BPA) treatment in female rats. Female rats were sc injected with BPA (50 or 500 µg/50 µl) or castor oil vehicle from postnatal day 1 to 10. At five months serum prolactin, pituitary GH, and serum and liver insulin growth factor-I (IGF-I) were measured by RIA. Major urinary proteins (MUPs) were determined by electrophoresis. Liver Cyp2c11, Cyp2c12, Adh1, Hnf6, and Prlr mRNA levels were determined by real time PCR. Pituitary GH content and liver IGF-I concentration were increased by neonatal BPA treatment, indicating partial masculinization of the GH axis in treated females. GH-dependent female predominant liver enzyme genes (Cyp2c12 and Adh1) and a transcription factor (Hnf6) were downregulated or defeminized, while there were no changes in a male predominant gene (Cyp2c11) or protein (MUP). Our findings indicate that perinatal exposure to BPA may compromise the sexually dimorphic capacity of the liver to metabolize drugs and steroids.


Assuntos
Disruptores Endócrinos/toxicidade , Estrogênios não Esteroides/toxicidade , Hormônio do Crescimento/metabolismo , Fígado/efeitos dos fármacos , Fenóis/toxicidade , Hipófise/efeitos dos fármacos , Fatores Etários , Envelhecimento/genética , Envelhecimento/metabolismo , Álcool Desidrogenase/genética , Animais , Animais Recém-Nascidos , Hidrocarboneto de Aril Hidroxilases/genética , Compostos Benzidrílicos , Família 2 do Citocromo P450 , Esquema de Medicação , Eletroforese em Gel de Poliacrilamida , Disruptores Endócrinos/administração & dosagem , Estrogênios não Esteroides/administração & dosagem , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Fator 6 Nuclear de Hepatócito/genética , Injeções Subcutâneas , Fator de Crescimento Insulin-Like I/metabolismo , Fígado/metabolismo , Masculino , Fenóis/administração & dosagem , Hipófise/metabolismo , Prolactina/sangue , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptores da Prolactina/genética , Caracteres Sexuais , Fatores Sexuais , Esteroide 16-alfa-Hidroxilase/genética , Esteroide Hidroxilases/genética
7.
Neuroendocrinology ; 92(4): 207-14, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20975260

RESUMO

Dopamine D2 receptor (D2R) participation in prolactin regulation is well documented, but the role of D2Rs in the control of other hormones involved in growth, food intake and glucose metabolism has not been extensively studied. The study of D2R knockout mice (Drd2(-/-)) puts forward new insights into the role of the D2R in growth hormone (GH)-releasing hormone-GH regulation, peptides involved in food intake, glucose homeostasis, as well as in prolactinoma development. The expected phenotype of chronic hyperprolactinemia and prolactinoma development was found in the Drd2(-/-) mouse, and this model constitutes a valuable tool in the study of dopamine-resistant prolactinomas. Unexpectedly, these mice were growth retarded, and the importance of functional hypothalamic D2Rs in the neonatal period was revealed. In the Drd2(-/-) mouse there was a failure of high neonatal GH levels and therefore the expansion of pituitary somatotropes was permanently altered. These mice also had increased food intake, and a sexually dimorphic participation of the D2R in food intake regulation is suggested. The effect described is probably secondary to D2R regulation of prolactin secretion. Furthermore, the negative modulation of D2Rs on α-melanocyte-stimulating hormone release and positive action on the hypothalamic expression of orexins reveals the complex D2R regulation of food intake. Finally, pancreatic D2Rs inhibit glucose-stimulated insulin release. Lack of dopaminergic inhibition throughout development in the Drd2(-/-) mouse may exert a gradual deteriorating effect on insulin homeostasis, so that eventually glucose intolerance develops. These results highlight the complex endocrine actions of the D2Rs at different levels, hypothalamus, pituitary or pancreas, which function to improve fitness, reproductive success and survival.


Assuntos
Sistema Endócrino/fisiologia , Metabolismo/genética , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/fisiologia , Animais , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Sistema Endócrino/metabolismo , Hormônio do Crescimento/metabolismo , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Camundongos , Camundongos Knockout , Prolactina/metabolismo , Receptores de Dopamina D2/metabolismo
8.
J Endocrinol ; 207(3): 301-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20943812

RESUMO

Abnormal exposure to steroid hormones within a critical developmental period elicits permanent alterations in female reproductive physiology in rodents, but the impact on the female GH axis and the underlying sexual differences in hepatic enzymes have not been described in detail. We have investigated the effect of neonatal androgenization of female mice (achieved by s.c. injection of 100 µg testosterone propionate (TP) on the day of birth: TP females) on the GHRH-somatostatin-GH axis and downstream GH targets, which included female and male predominant liver enzymes and secreted proteins. At 4 months of age, an organizational effect of neonatal testosterone was evidenced on hypothalamic Ghrh mRNA level but not on somatostatin (stt) mRNA level. Ghrh mRNA levels were higher in males than in females, but not in TP females. Increased expression in TP females correlated with increased pituitary GH content and somatotrope population, increased serum and liver IGF-I concentration, and ultimately higher body weight. Murine urinary proteins (MUPs) that were excreted at higher levels in male urine, and whose expression requires pulsatile occupancy of liver GH receptors, were not modified in TP females and neither was liver Mup 1/2/6/8 mRNA expression. Furthermore, a male predominant liver gene (Cyp2d9) was not masculinized in TP females either, whereas two female predominant genes (Cyp2b9 and Cyp2a4) were defeminized. These data support the hypothesis that neonatal steroid exposure contributes to the remodeling of the GH axis and defeminization of hepatic steroid-metabolizing enzymes, which may compromise liver physiology.


Assuntos
Hormônio do Crescimento/metabolismo , Fígado/metabolismo , Testosterona/metabolismo , Virilismo/metabolismo , Animais , Hidrocarboneto de Aril Hidroxilases/análise , Peso Corporal/fisiologia , Sistema Enzimático do Citocromo P-450/análise , Família 2 do Citocromo P450 , Feminino , Hormônio do Crescimento/análise , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Fator de Crescimento Insulin-Like I/análise , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Prolactina/sangue , Proteínas/análise , Esteroide Hidroxilases/análise , Testosterona/farmacologia , Virilismo/induzido quimicamente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA