Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 484
Filtrar
1.
Neuromolecular Med ; 26(1): 18, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38691185

RESUMO

Seipin is a key regulator of lipid metabolism, the deficiency of which leads to severe lipodystrophy. Hypothalamus is the pivotal center of brain that modulates appetite and energy homeostasis, where Seipin is abundantly expressed. Whether and how Seipin deficiency leads to systemic metabolic disorders via hypothalamus-involved energy metabolism dysregulation remains to be elucidated. In the present study, we demonstrated that Seipin-deficiency induced hypothalamic inflammation, reduction of anorexigenic pro-opiomelanocortin (POMC), and elevation of orexigenic agonist-related peptide (AgRP). Importantly, administration of rosiglitazone, a thiazolidinedione antidiabetic agent, rescued POMC and AgRP expression, suppressed hypothalamic inflammation, and restored energy homeostasis in Seipin knockout mice. Our findings offer crucial insights into the mechanism of Seipin deficiency-associated energy imbalance and indicates that rosiglitazone could serve as potential intervening agent towards metabolic disorders linked to Seipin.


Assuntos
Proteína Relacionada com Agouti , Metabolismo Energético , Subunidades gama da Proteína de Ligação ao GTP , Homeostase , Hipotálamo , Camundongos Knockout , Pró-Opiomelanocortina , Rosiglitazona , Animais , Camundongos , Hipotálamo/metabolismo , Metabolismo Energético/efeitos dos fármacos , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/biossíntese , Proteína Relacionada com Agouti/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Rosiglitazona/farmacologia , Masculino , Doenças Neuroinflamatórias/etiologia , Camundongos Endogâmicos C57BL , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Neuropeptídeos/genética , Neuropeptídeos/deficiência , Regulação da Expressão Gênica/efeitos dos fármacos
2.
Mol Psychiatry ; 28(3): 1365-1382, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36473997

RESUMO

Chronic stress exposure induces maladaptive behavioral responses and increases susceptibility to neuropsychiatric conditions. However, specific neuronal populations and circuits that are highly sensitive to stress and trigger maladaptive behavioral responses remain to be identified. Here we investigate the patterns of spontaneous activity of proopiomelanocortin (POMC) neurons in the arcuate nucleus (ARC) of the hypothalamus following exposure to chronic unpredictable stress (CUS) for 10 days, a stress paradigm used to induce behavioral deficits such as anhedonia and behavioral despair [1, 2]. CUS exposure increased spontaneous firing of POMC neurons in both male and female mice, attributable to reduced GABA-mediated synaptic inhibition and increased intrinsic neuronal excitability. While acute activation of POMC neurons failed to induce behavioral changes in non-stressed mice of both sexes, subacute (3 days) and chronic (10 days) repeated activation of POMC neurons was sufficient to induce anhedonia and behavioral despair in males but not females under non-stress conditions. Acute activation of POMC neurons promoted susceptibility to subthreshold unpredictable stress in both male and female mice. Conversely, acute inhibition of POMC neurons was sufficient to reverse CUS-induced anhedonia and behavioral despair in both sexes. Collectively, these results indicate that chronic stress induces both synaptic and intrinsic plasticity of POMC neurons, leading to neuronal hyperactivity. Our findings suggest that POMC neuron dysfunction drives chronic stress-related behavioral deficits.


Assuntos
Anedonia , Núcleo Arqueado do Hipotálamo , Depressão , Neurônios , Pró-Opiomelanocortina , Estresse Psicológico , Animais , Feminino , Masculino , Camundongos , Doença Aguda , Anedonia/fisiologia , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/fisiopatologia , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , Doença Crônica , Excitabilidade Cortical/fisiologia , Depressão/metabolismo , Depressão/fisiopatologia , Modelos Animais de Doenças , Transtornos Mentais/metabolismo , Transtornos Mentais/fisiopatologia , Camundongos Endogâmicos C57BL , Fenômenos Fisiológicos do Sistema Nervoso , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Sinapses/metabolismo , Sinapses/fisiologia
3.
Mol Biol Rep ; 47(10): 8273-8278, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32914264

RESUMO

Neuropeptides are important, multifunctional regulatory factors of the nervous system, being considered as a novel, atypical sites of antidepressants action. It has already been proven that some of them, such as selective serotonin reuptake inhibitors (SSRI), are able to affect peptidergic pathways in various brain regions. Despite these reports, there is so far no reports regarding the effect of treatment with SSRIs on brain proopiomelanocortin (POMC), kisspeptin, Kiss1R and MCHR1 gene expression. In the current study we examined POMC, kisspeptin, Kiss1R and MCHR1 mRNA expression in the selected brain structures (hypothalamus, hippocampus, amygdala, striatum, cerebellum and brainstem) of rats chronically treated with a 10 mg/kg dose of escitalopram using quantitative Real-Time PCR. Long-term treatment with escitalopram led to the upregulation of MCHR1 expression in the rat amygdala. Kisspeptin mRNA level was also increased in the amygdala, but Kiss1R mRNA expressions were elevated in the hippocampus, hypothalamus and cerebellum. POMC mRNA expressions were in turn decreased in the hippocampus, amygdala, cerebellum and brainstem. These results may support the hypothesis that these neuropeptides may be involved in the site-dependent actions of SSRI antidepressants. This is the first report of the effects of escitalopram on POMC, kisspeptin, Kiss1R and MCHR1 in animal brain. Our findings shed a new light on the pharmacology of SSRIs and may contribute to a better understanding of the alternative, neuropeptide-dependent modes of antidepressant action.


Assuntos
Encéfalo/metabolismo , Citalopram/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Kisspeptinas/biossíntese , Pró-Opiomelanocortina/biossíntese , Receptores de Kisspeptina-1/biossíntese , Receptores de Somatostatina/biossíntese , Animais , Masculino , Ratos , Ratos Sprague-Dawley
4.
Mol Biol Rep ; 47(8): 5953-5962, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32705506

RESUMO

Pro-opiomelanocortin (POMC) is a large precursor protein of and ß-endorphin. POMC expressed in keratinocytes regulates various pathophysiological responses, such as pruritus in atopic dermatitis. Interleukin (IL)-31 is a T helper 2 (Th2)-derived cytokine that functions as a pruritogen, stimulating the sensory neurons in the skin. However, the regulatory mechanism underlying IL-31-induced POMC expression in keratinocytes remains largely unknown. Herein, using a 5'-serial deletion and site-specific mutation constructs of the regulatory region of POMC, we demonstrated that a putative EGR1-binding sequence (EBS) motif in POMC is required for its upregulation by IL-31 in HaCaT keratinocytes. Notably, EGR-1 directly interacted with the EBS motif in POMC. The ectopic expression of EGR-1 stimulated the POMC promoter activity, whereas the knockdown of EGR-1 expression by RNA interference reduced IL-31-induced POMC expression. Furthermore, we observed that three major mitogen-activated protein kinases, ERK, JNK, and p38 kinase, mediated IL-31-induced EGR-1 expression. In summary, our results suggest that EGR-1 trans-activates POMC in response to IL-31 stimulation in HaCaT keratinocytes.


Assuntos
Proteína 1 de Resposta de Crescimento Precoce/fisiologia , Interleucinas/farmacologia , Queratinócitos/metabolismo , Pró-Opiomelanocortina/genética , Transcrição Gênica/efeitos dos fármacos , Motivos de Aminoácidos , Linhagem Celular Transformada , Proteína 1 de Resposta de Crescimento Precoce/antagonistas & inibidores , Proteína 1 de Resposta de Crescimento Precoce/genética , Genes Reporter , Genes Sintéticos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Mutação Puntual , Pró-Opiomelanocortina/biossíntese , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Ativação Transcricional , Regulação para Cima/efeitos dos fármacos
5.
Nutr. hosp ; 36(6): 1354-1360, nov.-dic. 2019. tab, graf
Artigo em Inglês | IBECS | ID: ibc-191156

RESUMO

Background: folic acid participates in one-carbon metabolism, which supplies methyl groups to numerous reactions in the body. Impaired delivery of these methyl groups affects gene expression. We hypothesize that offspring exposed to less folic acid will express higher levels of Pomc (proopiomelanocortin) gene mRNA. Aim: to investigate the Pomc gene and protein expression pattern in the female offspring of female rats receiving a folic acid-deficient diet during gestation, lactation, and post-weaning. Methods: the study involved female rat offspring (n = 10) born from mothers subjected to a control (2.0 mg of folic acid/kg of food) or folic acid-deficient (0.5 mg of folic acid/kg of food) diet, and fed the same diet during post-weaning. Samples were collected from the arcuate nucleus of the hypothalamus of the female offspring for real-time PCR and Western blotting analyses. Results: the female offspring in the folic acid-deficient diet group had significantly higher Pomc gene and protein expression than the female offspring in the control diet group (p = 0.03, p = 0.01, respectively). Conclusion: a folic acid-deficient diet during gestation, lactation, and post-weaning increases Pomc gene and protein expression, but does not modify food intake or body weight of female rat offspring


Antecedentes: el ácido fólico participa en el metabolismo de un solo carbono, que suministra grupos metilo a numerosas reacciones del cuerpo. La aportación alterada de estos grupos metilo afecta a la expresión génica. Nuestra hipótesis es que la descendencia expuesta a menos ácido fólico expresará niveles más altos de ARNm del gen Pomc (proopiomelanocortina). Objetivo: investigar el patrón de expresión del gen Pomc y de sus proteínas en crías de ratas hembras que recibieron una dieta deficiente en ácido fólico durante la gestación, la lactancia y el destete. Métodos: el estudio incluyó crías hembras (n = 10) nacidas de madres sometidas a una dieta control (2,0 mg de ácido fólico/kg de alimento) o deficiente en ácido fólico (0,5 mg de ácido fólico/kg de alimento) durante la gestación y la lactancia, y alimentadas con la misma dieta durante el destete. Se recolectaron muestras del núcleo arqueado del hipotálamo de las hembras para el análisis de la expresión génica (PCR en tiempo real) y de proteínas (inmunomanchado Western). Resultados: las hembras pertenecientes al grupo de dieta deficiente en ácido fólico tuvieron una expresión del gen Pomc y sus proteínas significativamente mayor que la de las hembras pertenecientes al grupo con dieta de control (p = 0,03, p = 0,01, respectivamente). Conclusión: la dieta deficiente en ácido fólico durante la gestación, la lactancia y el destete modifica la expresión del gen Pomc y sus proteínas pero no modifica la ingesta de alimentos ni el peso corporal de las ratas hembra


Assuntos
Animais , Feminino , Gravidez , Ratos , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/genética , Desmame , Dieta , Ácido Fólico/administração & dosagem , Animais Recém-Nascidos , Expressão Gênica , Ratos Wistar
6.
Nutr Hosp ; 36(6): 1354-1360, 2019 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-31691569

RESUMO

INTRODUCTION: Background: folic acid participates in one-carbon metabolism, which supplies methyl groups to numerous reactions in the body. Impaired delivery of these methyl groups affects gene expression. We hypothesize that offspring exposed to less folic acid will express higher levels of Pomc (proopiomelanocortin) gene mRNA. Aim: to investigate the Pomc gene and protein expression pattern in the female offspring of female rats receiving a folic acid-deficient diet during gestation, lactation, and post-weaning. Methods: the study involved female rat offspring (n = 10) born from mothers subjected to a control (2.0 mg of folic acid/kg of food) or folic acid-deficient (0.5 mg of folic acid/kg of food) diet, and fed the same diet during post-weaning. Samples were collected from the arcuate nucleus of the hypothalamus of the female offspring for real-time PCR and Western blotting analyses. Results: the female offspring in the folic acid-deficient diet group had significantly higher Pomc gene and protein expression than the female offspring in the control diet group (p = 0.03, p = 0.01, respectively). Conclusion: a folic acid-deficient diet during gestation, lactation, and post-weaning increases Pomc gene and protein expression, but does not modify food intake or body weight of female rat offspring.


INTRODUCCIÓN: Antecedentes: el ácido fólico participa en el metabolismo de un solo carbono, que suministra grupos metilo a numerosas reacciones del cuerpo. La aportación alterada de estos grupos metilo afecta a la expresión génica. Nuestra hipótesis es que la descendencia expuesta a menos ácido fólico expresará niveles más altos de ARNm del gen Pomc (proopiomelanocortina). Objetivo: investigar el patrón de expresión del gen Pomc y de sus proteínas en crías de ratas hembras que recibieron una dieta deficiente en ácido fólico durante la gestación, la lactancia y el destete. Métodos: el estudio incluyó crías hembras (n = 10) nacidas de madres sometidas a una dieta control (2,0 mg de ácido fólico/kg de alimento) o deficiente en ácido fólico (0,5 mg de ácido fólico/kg de alimento) durante la gestación y la lactancia, y alimentadas con la misma dieta durante el destete. Se recolectaron muestras del núcleo arqueado del hipotálamo de las hembras para el análisis de la expresión génica (PCR en tiempo real) y de proteínas (inmunomanchado Western). Resultados: las hembras pertenecientes al grupo de dieta deficiente en ácido fólico tuvieron una expresión del gen Pomc y sus proteínas significativamente mayor que la de las hembras pertenecientes al grupo con dieta de control (p = 0,03, p = 0,01, respectivamente). Conclusión: la dieta deficiente en ácido fólico durante la gestación, la lactancia y el destete modifica la expresión del gen Pomc y sus proteínas pero no modifica la ingesta de alimentos ni el peso corporal de las ratas hembra.


Assuntos
Dieta , Ácido Fólico/administração & dosagem , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/genética , Desmame , Animais , Animais Recém-Nascidos , Feminino , Expressão Gênica , Gravidez , Ratos , Ratos Wistar
7.
Physiol Behav ; 210: 112644, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31398442

RESUMO

Several studies in rodents and layer chickens have demonstrated that insulin upregulates hypothalamic AKT-mediated signaling and expression of proopiomelanocortin (POMC, the precursor of alpha-melanocyte stimulating hormone, an anorexigenic peptide) and suppresses appetite in these animals. However, a previous study has also reported that insulin fails to suppress food intake in broiler chicks. In the present study, no significant differences were observed in hypothalamic AKT and forkhead box O1 (FOXO1) phosphorylation levels between broiler and layer chicks. The phosphorylation rate of AKT, but not that of FOXO1, increased in the hypothalami of broilers refed for 1 h after a 24-h fast, with a corresponding increase in plasma insulin concentration. Intracerebroventricular (ICV) administration of 50 pmol insulin, which could decrease food intake in broiler chicks, significantly increased the AKT phosphorylation rate, whereas no significant change was observed in FOXO1 phosphorylation or POMC expression after ICV insulin administration. These findings suggest that hypothalamic AKT responds to insulin in broiler chicks, but FOXO1-mediated regulation of POMC expression is not induced by insulin, which may be one of the causes of excessive food intake in broiler chickens.


Assuntos
Galinhas , Ingestão de Alimentos , Proteína Forkhead Box O1/metabolismo , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Proteína Oncogênica v-akt/metabolismo , Animais , Animais Recém-Nascidos , Hipoglicemiantes/administração & dosagem , Injeções Intraventriculares , Insulina/administração & dosagem , Insulina/sangue , Masculino , Fosforilação , Pró-Opiomelanocortina/biossíntese , Transdução de Sinais/efeitos dos fármacos
8.
Neurosci Lett ; 711: 134407, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31369799

RESUMO

Glucocorticoids and glucocorticoid receptors (GRs) suppress pituitary pro-opiomelanocortin (Pomc) gene expression. O-linked ß-N-acetylglucosamine (O-GlcNAc) modification, mediated by O-GlcNAc transferase (OGT), plays an important role during gene transcription. However, whether OGT is involved in the GR-mediated transrepression that occurs in pituitary corticotroph cells is currently unknown. Here, we report that OGT regulates Pomc expression in the mouse corticotroph cell line AtT-20. The overexpression of OGT has an additive effect on the GR-mediated negative transcription pathway. Both the knockdown of OGT by RNA interference and the use of a chemical OGT inhibitor abolished the repressive effects of Pomc expression induced by GRs. OGT inhibition leads to both the decreased recruitment of GRs and the increased recruitment of RNA polymerase II to the Pomc locus. O-GlcNAc modification is involved in the negative regulation of Pomc transcription in corticotroph cells. OGT may be a promising therapeutic target for the treatment of Cushing's disease.


Assuntos
Corticotrofos/metabolismo , Regulação da Expressão Gênica/fisiologia , N-Acetilglucosaminiltransferases/metabolismo , Pró-Opiomelanocortina/biossíntese , Animais , Linhagem Celular , Camundongos
9.
Neuroendocrinology ; 109(4): 362-373, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30995664

RESUMO

BACKGROUND: Cushing's disease (CD) is caused by adrenocorticotropic hormone (ACTH)-secreting pituitary tumours. They express high levels of heat shock protein 90 and heat shock factor 1 (HSF1) in comparison to the normal tissue counterpart, indicating activated cellular stress. AIMS: Our objectives were: (1) to correlate HSF1 expression with clinical features and hormonal/radiological findings of CD, and (2) to investigate the effects of HSF1 inhibition as a target for CD treatment. PATIENTS/METHODS: We examined the expression of total and pSer326HSF1 (marker for its transcriptional activation) by Western blot on eight human CD tumours and compared to the HSF1 status of normal pituitary. We screened a cohort of 45 patients with CD for HSF1 by immunohistochemistry and correlated the HSF1 immunoreactivity score with the available clinical data. We evaluated the effects of HSF1 silencing with RNA interference and the HSF1 inhibitor KRIBB11 in AtT-20 cells and four primary cultures of human corticotroph tumours. RESULTS: We show that HSF1 protein is highly expressed and transcriptionally active in CD tumours in comparison to normal pituitary. The immunoreactivity score for HSF1 did not correlate with the typical clinical features of the disease. HSF1 inhibition reduced proopiomelanocortin (Pomc) transcription in AtT-20 cells. The HSF1 inhibitor KRIBB11 suppressed ACTH synthesis from 75% of human CD tumours in primary cell culture. This inhibitory action on Pomc transcription was mediated by increased glucocorticoid receptor and suppressed Nurr77/Nurr1 and AP-1 transcriptional activities. CONCLUSIONS: These data show that HSF1 regulates POMC transcription. Pharmacological targeting of HSF1 may be a promising treatment option for the control of excess ACTH secretion in CD.


Assuntos
Fatores de Transcrição de Choque Térmico/antagonistas & inibidores , Hipersecreção Hipofisária de ACTH/tratamento farmacológico , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/genética , Hormônio Adrenocorticotrópico/biossíntese , Adulto , Aminopiridinas/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Inativação Gênica , Fatores de Transcrição de Choque Térmico/genética , Humanos , Imuno-Histoquímica , Indazóis/farmacologia , Masculino , Hipersecreção Hipofisária de ACTH/metabolismo , Interferência de RNA , Fator de Transcrição AP-1/farmacologia , Ativação Transcricional/efeitos dos fármacos , Adulto Jovem
10.
Biochem J ; 476(5): 827-842, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30787050

RESUMO

To closely mimic physiological conditions, low oxygen cultures have been employed in stem cell and cancer research. Although in vivo oxygen concentrations in tissues are often much lower than ambient 21% O2 (ranging from 3.6 to 12.8% O2), most cell cultures are maintained at 21% O2 To clarify the effects of the O2 culture concentration on the regulated secretion of peptide hormones in neuro-endocrine cells, we examined the changes in the storage and release of peptide hormones in neuro-endocrine cell lines and endocrine tissues cultured in a relatively lower O2 concentration. In both AtT-20 cells derived from the mouse anterior pituitary and freshly prepared mouse pituitaries cultured in 10% O2 for 24 h, the storage and regulated secretion of the mature peptide hormone adrenocorticotropic hormone were significantly increased compared with those in cells and pituitaries cultured in ambient 21% O2, whereas its precursor proopiomelanocortin was not increased in the cells and tissues after being cultured in 10% O2 Simultaneously, the prohormone-processing enzymes PC1/3 and carboxypeptidase E were up-regulated in cells cultured in 10% O2, thus facilitating the conversion of prohormones to their active form. Similarly, culturing the mouse ß-cell line MIN6 and islet tissue in 10% O2 also significantly increased the conversion of proinsulin into mature insulin, which was secreted in a regulated manner. These results suggest that culture under 10% O2 is more optimal for endocrine tissues/cells to efficiently generate and secrete active peptide hormones than ambient 21% O2.


Assuntos
Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Células Neuroendócrinas/metabolismo , Oxigênio/farmacologia , Adeno-Hipófise/metabolismo , Pró-Opiomelanocortina/biossíntese , Regulação para Cima/efeitos dos fármacos , Animais , Técnicas de Cultura de Células , Linhagem Celular , Camundongos
11.
J Biol Chem ; 294(13): 4946-4955, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30709906

RESUMO

Hypoxia-inducible factor-2α (HIF2α) is a nuclear transcription factor that plays a critical role in cell survival including metabolic adaptation under hypoxia as well as normoxia, but whether HIF2α contributes to the control of whole-body metabolic balance is unclear. In this study, we found that the hypothalamic HIF2α protein level rapidly increases in young mice that are centrally stimulated with insulin. However, this insulin-induced HIF2α up-regulation is substantially attenuated in mice of advanced age. This attenuation is comparable with the effect of high-calorie feeding in young mice. Of note, unlike high-calorie feeding conditions, age-dependent HIF2α attenuation occurs without impaired activation of the hypothalamic IR/IRS-2/AKT/FOXO1 pathway in response to insulin. Molecular and physiological analyses revealed that hypothalamic HIF2α contributes to the action of central insulin in regulation of proopiomelanocortin (Pomc) gene expression and food intake. HIF2α knockout in POMC neurons led to age-dependent excess weight gain and fat increase, a phenotype that was associated with a mild degree of glucose intolerance and insulin resistance. In conclusion, hypothalamic HIF2α responds to insulin, and the up-regulation is involved in adaptive metabolic regulation as age increases, whereas impairment of HIF2α in the hypothalamus contributes to weight gain and glucose disorders in age-dependent manners.


Assuntos
Envelhecimento/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Intolerância à Glucose/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Transdução de Sinais , Envelhecimento/genética , Envelhecimento/patologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Regulação da Expressão Gênica , Intolerância à Glucose/genética , Intolerância à Glucose/patologia , Hipotálamo/patologia , Insulina/genética , Camundongos , Camundongos Transgênicos , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/genética
12.
Elife ; 72018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30311908

RESUMO

Proopiomelanocortin (POMC) neurons are major negative regulators of energy balance. A distinct developmental property of POMC neurons is that they can adopt an orexigenic neuropeptide Y (NPY) phenotype. However, the mechanisms underlying the differentiation of Pomc progenitors remain unknown. Here, we show that the loss of the microRNA (miRNA)-processing enzyme Dicer in POMC neurons causes metabolic defects, an age-dependent decline in the number of PomcmRNA-expressing cells, and an increased proportion of Pomc progenitors acquiring a NPY phenotype. miRNome microarray screening further identified miR-103/107 as candidates that may be involved in the maturation of Pomc progenitors. In vitro inhibition of miR-103/107 causes a reduction in the number of Pomc-expressing cells and increases the proportion of Pomc progenitors differentiating into NPY neurons. Moreover, in utero silencing of miR-103/107 causes perturbations in glucose homeostasis. Together, these data suggest a role for prenatal miR-103/107 in the maturation of Pomc progenitors and glucose homeostasis.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/metabolismo , Neurônios/fisiologia , Neuropeptídeo Y/biossíntese , Pró-Opiomelanocortina/biossíntese , Animais , Glucose/metabolismo , Homeostase , Camundongos
13.
J Comp Neurol ; 526(15): 2444-2461, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30242838

RESUMO

We recently reported that the number of hypothalamic tanycytes expressing pro-opiomelanocortin (Pomc) is highly variable among brains of adult rats. While its cause and significance remain unknown, identifying other variably expressed genes in tanycytes may help understand this curious phenomenon. In this in situ hybridization study, we report that the Prss56 gene, which encodes a trypsin-like serine protease and is expressed in neural stem/progenitor cells, shows a similarly variable mRNA expression in tanycytes of adult rats and correlates inversely with tanycyte Pomc mRNA. Prss56 was expressed in α1, ß1, subsets of α2, and some median eminence γ tanycytes, but virtually absent from ß2 tanycytes. Prss56 was also expressed in vimentin positive tanycyte-like cells in the parenchyma of the ventromedial and arcuate nuclei, and in thyrotropin beta subunit-expressing cells of the pars tuberalis of the pituitary. In contrast to adults, Prss56 expression was uniformly high in tanycytes in adolescent rats. In mice, Prss56-expressing tanycytes and parenchymal cells were also observed but fewer in number and without significant variations. The results identify Prss56 as a second gene that is expressed variably in tanycytes of adult rats. We propose that the variable, inversely correlating expression of Prss56 and Pomc reflect periodically oscillating gene expression in tanycytes rather than stable expression levels that vary between individual rats. A possible functional link between Prss56 and POMC, and Prss56 as a potential marker for migrating tanycytes are discussed.


Assuntos
Células Ependimogliais/metabolismo , Hipotálamo/metabolismo , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/genética , Serina Proteases/biossíntese , Serina Proteases/genética , Envelhecimento/metabolismo , Animais , Contagem de Células , Células Ependimogliais/classificação , Feminino , Regulação da Expressão Gênica , Hipotálamo/química , Antígeno Ki-67/metabolismo , Masculino , Hipófise/metabolismo , Ratos , Ratos Sprague-Dawley , Serina Proteases/metabolismo , Terminologia como Assunto , Tireotropina/biossíntese , Tireotropina/genética
14.
Proc Natl Acad Sci U S A ; 115(40): E9489-E9498, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30224492

RESUMO

Two classes of peptide-producing neurons in the arcuate nucleus (Arc) of the hypothalamus are known to exert opposing actions on feeding: the anorexigenic neurons that express proopiomelanocortin (POMC) and the orexigenic neurons that express agouti-related protein (AgRP) and neuropeptide Y (NPY). These neurons are thought to arise from a common embryonic progenitor, but our anatomical and functional understanding of the interplay of these two peptidergic systems that contribute to the control of feeding remains incomplete. The present study uses a combination of optogenetic stimulation with viral and transgenic approaches, coupled with neural activity mapping and brain transparency visualization to demonstrate the following: (i) selective activation of Arc POMC neurons inhibits food consumption rapidly in unsated animals; (ii) activation of Arc neurons arising from POMC-expressing progenitors, including POMC and a subset of AgRP neurons, triggers robust feeding behavior, even in the face of satiety signals from POMC neurons; (iii) the opposing effects on food intake are associated with distinct neuronal projection and activation patterns of adult hypothalamic POMC neurons versus Arc neurons derived from POMC-expressing lineages; and (iv) the increased food intake following the activation of orexigenic neurons derived from POMC-expressing progenitors engages an extensive neural network that involves the endogenous opioid system. Together, these findings shed further light on the dynamic balance between two peptidergic systems in the moment-to-moment regulation of feeding behavior.


Assuntos
Proteína Agouti Sinalizadora/biossíntese , Núcleo Arqueado do Hipotálamo/metabolismo , Comportamento Alimentar/fisiologia , Neurônios/metabolismo , Neuropeptídeo Y/biossíntese , Pró-Opiomelanocortina/biossíntese , Transdução de Sinais/fisiologia , Proteína Agouti Sinalizadora/genética , Animais , Núcleo Arqueado do Hipotálamo/citologia , Comportamento Alimentar/psicologia , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Neuropeptídeo Y/genética , Pró-Opiomelanocortina/genética
15.
BMC Anesthesiol ; 18(1): 96, 2018 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-30053804

RESUMO

BACKGROUND: Several hypnotic drugs have been previously identified as modulators of food intake, but exact mechanisms remain unknown. Feeding behavior implicates several neuronal populations in the hypothalamic arcuate nucleus including orexigenic neuropeptide Y and anorexigenic pro-opiomelanocortin producing neurons. The aim of this study was to investigate in mice the impact of different hypnotic drugs on food consumption and neuropeptide Y or pro-opiomelanocortine mRNA expression level in the hypothalamic arcuate nucleus. METHODS: Saline control, isoflurane, thiopental, midazolam or propofol were administered to C57Bl/6 mice. Feeding behavior was evaluated during 6 h. In situ hybridization of neuropeptide Y and pro-opiomelanocortine mRNAs in the hypothalamus brain region was also performed. Data were analyzed by Kruskal Wallis test and analysis of variance (p < 0.05). RESULTS: Midazolam, thiopental and propofol induced feeding behavior. Midazolam and thiopental increased neuropeptide Y mRNA level (respectively by 106 and 125%, p < 0.001) compared with control. Propofol and midazolam decreased pro-opiomelanocortine mRNA level by 31% (p < 0,01) compared with control. Isoflurane increased pro-opiomelanocortine mRNA level by 40% compared with control. CONCLUSION: In our murine model, most hypnotics induced food consumption. The hypnotic-induced regulation of neuropeptide Y and pro-opiomelanocortine hypothalamic peptides is associated with this finding. Our data suggest that administration of some hypnotic drugs may affect hypothalamic peptide precursor and neuropeptide expression and concomittantly modulate food intake. Thus, this questions the choice of anesthetics for better care management of patients undergoing major surgery or at risk of undernutrition.


Assuntos
Anestésicos/farmacologia , Núcleo Arqueado do Hipotálamo/metabolismo , Comportamento Alimentar/efeitos dos fármacos , Neuropeptídeo Y/biossíntese , Pró-Opiomelanocortina/biossíntese , Animais , Masculino , Camundongos
16.
Pharmacol Rep ; 70(4): 650-657, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29902669

RESUMO

BACKGROUND: The endocannabinoid (eCB) system is strongly involved in the regulation of anxiety and feeding behavior. RVD-hemopressin(α) [RVD-hp(α)], a N-terminally extended form of hemopressin, is a negative allosteric modulator of the cannabinoid (CB) 1 receptor and a positive allosteric modulator of CB2 receptor which has been recently reported to exert anxiolytic/antidepressant and anorexigenic effects after peripheral administration in rats. Pharmacological evidences reported a possible link between brain hypocretin/orexin, monoamine and eCB systems, as regards appetite and emotional behavior control. Considering this, the aim of our work was to investigated the effects of RVD-hp(α) on anxiety like behavior and food intake after central administration and related it to monoamine levels and orexin-A gene expression, in the hypothalamus. METHODS: We have studied the effects of central RVD-hp(α) (10nmol) injection on anxiety-like behavior and feeding using different behavioral tests. Hypothalamic levels of norepinephrine (NE), dopamine (DA) and serotonin (5-hydroxytryptamine, 5-HT) and gene expression of orexin-A and proopiomelanocortin (POMC) were measured by high performance liquid chromatography (HPLC) and real-time reverse transcription polymerase chain reaction (RT-PCR) analysis, respectively. RESULTS: Central RVD-hp(α) administration decreased locomotion activity and stereotypies. Moreover, RVD-hp(α) treatment inhibited anxiogenic-like behavior and food intake, NE levels and orexin-A gene expression, in the hypothalamus. CONCLUSION: Concluding, in the present study we demonstrated that central RVD-hp(α) induced anxiolytic and anorexigenic effects possibly related to reduced NE and orexin-A and POMC signaling, in the hypothalamus. These findings further support the central role of the peptide in rat brain thus representing an innovative pharmacological approach for designing new anorexigenic drugs targeting eCB system.


Assuntos
Comportamento Exploratório/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Hemoglobinas/farmacologia , Hipotálamo/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Norepinefrina/metabolismo , Orexinas/biossíntese , Fragmentos de Peptídeos/farmacologia , Animais , Dopamina/metabolismo , Hemoglobinas/administração & dosagem , Hipotálamo/metabolismo , Injeções Intraventriculares , Masculino , Atividade Motora/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Pró-Opiomelanocortina/biossíntese , Ratos , Serotonina/metabolismo , Comportamento Estereotipado/efeitos dos fármacos
17.
J Neural Transm (Vienna) ; 125(7): 1099-1105, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29488100

RESUMO

Neuroleptics modulate the expression level of some regulatory neuropeptides in the brain. However, if these therapeutics influence the peptidergic circuits in the amygdala remains unclear. This study specifies the impact profile of the classical antipsychotic drugs on mRNA expression of the spexin/NPQ, kisspeptin-1 and POMC in the rat amygdala. Animals were treated with haloperidol and chlorpromazine for 28 days prior to transcript quantification via qPCR. Haloperidol and chlorpromazine induced a change in the expression of all neuropeptides analyzed. Both drugs led to the decrease of Kiss-1 expression, whereas in POMC and spexin/NPQ their up-regulation in the amygdala was detected. These modulating effects on may represent alternative, so far unknown mechanisms, of classical antipsychotic drugs triggering pharmacological responses.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Antipsicóticos/farmacologia , Kisspeptinas/efeitos dos fármacos , Hormônios Peptídicos/efeitos dos fármacos , Pró-Opiomelanocortina/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Expressão Gênica/efeitos dos fármacos , Kisspeptinas/biossíntese , Masculino , Hormônios Peptídicos/biossíntese , Pró-Opiomelanocortina/biossíntese , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley
18.
Neurochem Res ; 43(4): 821-837, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29397535

RESUMO

The pro-opiomelanocortin (POMC)-expressing neurons of the hypothalamic arcuate nucleus (ARC) are involved in the control of food intake and metabolic processes. It is assumed that, in addition to leptin, the activity of these neurons is regulated by serotonin and dopamine, but only subtype 2C serotonin receptors (5-HT2CR) was identified earlier on the POMC-neurons. The aim of this work was a comparative study of the localization and number of leptin receptors (LepR), types 1 and 2 dopamine receptors (D1R, D2R), 5-HT1BR and 5-HT2CR on the POMC-neurons and the expression of the genes encoding them in the ARC of the normal and diet-induced obese (DIO) rodents and the agouti mice (A y /a) with the melanocortin obesity. As shown by immunohistochemistry (IHC), all the studied receptors were located on the POMC-immunopositive neurons, and their IHC-content was in agreement with the expression of their genes. In DIO rats the number of D1R and D2R in the POMC-neurons and their expression in the ARC were reduced. In DIO mice the number of D1R and D2R did not change, while the number of LepR and 5-HT2CR was increased, although to a small extent. In the POMC-neurons of agouti mice the number of LepR, D2R, 5-HT1BR and 5-HT2CR was increased, and the D1R number was reduced. Thus, our data demonstrates for the first time the localization of different types of the serotonin and dopamine receptors on the POMC-neurons and a specific pattern of the changes of their number and expression in the DIO and melanocortin obesity.


Assuntos
Hipotálamo/metabolismo , Obesidade/metabolismo , Pró-Opiomelanocortina/biossíntese , Receptores Dopaminérgicos/biossíntese , Receptores para Leptina/biossíntese , Receptores de Serotonina/biossíntese , Animais , Feminino , Hipotálamo/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/química , Neurônios/metabolismo , Pró-Opiomelanocortina/análise , Ratos , Ratos Wistar , Receptores Dopaminérgicos/análise , Receptores para Leptina/análise , Receptores de Serotonina/análise , Roedores
19.
Mol Cell Biochem ; 447(1-2): 165-174, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29392534

RESUMO

"With no lysine" (WNK) kinases have been shown to regulate various ion transporters in various tissues, but studies on the function of WNK kinases in the brain have been limited. In this study, we discovered that WNK1 and WNK4 in POMC-expressing neuronal cells in WNK1 overexpressed transgenic mice (WNK1 TG) decrease appetite via degradation of Kir6.2. Weight gain after 20 weeks of age was delayed in WNK1 TG mice as a result of reduced food intake. Expression of WNK1 and proopiomelanocortin (POMC) was higher in POMC-expressing neurons in the hypothalamus of WNK1 TG mice than in WT mice. Immunostaining of serial sections of the hypothalamus revealed that POMC-expressing neurons were smaller in WNK1 TG mice than in WT mice. In addition, expression of Kir6.2 was significantly reduced in WNK1 TG mice. Overexpression and knockdown of WNK4 demonstrated that WNK4 regulates protein expression of Kir6.2 via protein-protein interaction. Accordingly, reduced age-dependent weight gain of WNK1 TG mice seems to be related with the decreased Kir6.2 expression via WNK1- and WNK4-regulated protein stability of Kir6.2.


Assuntos
Regulação da Expressão Gênica , Hipotálamo/metabolismo , Canais KATP/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Proteólise , Proteína Quinase 1 Deficiente de Lisina WNK/biossíntese , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Células HEK293 , Humanos , Hipotálamo/citologia , Canais KATP/genética , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Pró-Opiomelanocortina/genética , Proteínas Serina-Treonina Quinases/genética , Estabilidade Proteica , Ratos , Proteína Quinase 1 Deficiente de Lisina WNK/genética
20.
Proc Natl Acad Sci U S A ; 114(31): 8396-8401, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28716934

RESUMO

Sigma-1 antagonism potentiates the antinociceptive effects of opioid drugs, so sigma-1 receptors constitute a biological brake to opioid drug-induced analgesia. The pathophysiological role of this process is unknown. We aimed to investigate whether sigma-1 antagonism reduces inflammatory pain through the disinhibition of the endogenous opioidergic system in mice. The selective sigma-1 antagonists BD-1063 and S1RA abolished mechanical and thermal hyperalgesia in mice with carrageenan-induced acute (3 h) inflammation. Sigma-1-mediated antihyperalgesia was reversed by the opioid antagonists naloxone and naloxone methiodide (a peripherally restricted naloxone analog) and by local administration at the inflamed site of monoclonal antibody 3-E7, which recognizes the pan-opioid sequence Tyr-Gly-Gly-Phe at the N terminus of most endogenous opioid peptides (EOPs). Neutrophils expressed pro-opiomelanocortin, the precursor of ß-endorphin (a known EOP), and constituted the majority of the acute immune infiltrate. ß-endorphin levels increased in the inflamed paw, and this increase and the antihyperalgesic effects of sigma-1 antagonism were abolished by reducing the neutrophil load with in vivo administration of an anti-Ly6G antibody. The opioid-dependent sigma-1 antihyperalgesic effects were preserved 5 d after carrageenan administration, where macrophages/monocytes were found to express pro-opiomelanocortin and to now constitute the majority of the immune infiltrate. These results suggest that immune cells harboring EOPs are needed for the antihyperalgesic effects of sigma-1 antagonism during inflammation. In conclusion, sigma-1 receptors curtail immune-driven peripheral opioid analgesia, and sigma-1 antagonism produces local opioid analgesia by enhancing the action of EOPs of immune origin, maximizing the analgesic potential of immune cells that naturally accumulate in painful inflamed areas.


Assuntos
Analgesia/métodos , Analgésicos Opioides/farmacologia , Morfolinas/farmacologia , Naloxona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Receptores sigma/antagonistas & inibidores , Animais , Antígenos Ly/imunologia , Carragenina/toxicidade , Feminino , Inflamação/tratamento farmacológico , Inflamação/patologia , Macrófagos/metabolismo , Camundongos , Naloxona/farmacologia , Neutrófilos/metabolismo , Oligopeptídeos/metabolismo , Dor/tratamento farmacológico , Piperazinas/farmacologia , Pró-Opiomelanocortina/biossíntese , Pirazóis/farmacologia , Compostos de Amônio Quaternário/farmacologia , Receptores sigma/metabolismo , Receptor Sigma-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...