Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroendocrinology ; 111(10): 986-997, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33152734

RESUMO

Glucagon-like peptide-1 (GLP-1) exerts its anorexigenic effect at least partly via the proopiomelanocortin (POMC) neurons of the arcuate (ARC) nucleus. These neurons are known to express GLP-1 receptor (GLP-1R). The aim of the study was to determine whether in addition to its direct effect, GLP-1 also modulates how neuronal inputs can regulate the POMC neurons by acting on presynaptic terminals, ultrastructural and electrophysiological studies were performed on tissues of adult male mice. GLP-1R-immunoreactivity was associated with the cell membrane of POMC neurons and with axon terminals forming synapses on these cells. The GLP-1 analog exendin 4 (Ex4) markedly increased the firing rate of all examined POMC neurons and depolarized these cells. These effects of Ex4 were prevented by intracellular administration of the G-protein blocker guanosine 5'-[ß-thio]diphosphate trilithium salt (GDP-ß-S). Ex4 also influenced the miniature postsynaptic currents (mPSCs) and evoked PSCs of POMC neurons. Ex4 increased the frequency of miniature excitatory PSCs (EPSCs) and the amplitude of the evoked EPSCs in half of the POMC neurons. Ex4 increased the frequency of miniature inhibitory PSCs (IPSCs) and the amplitudes of the evoked IPSCs in one-third of neurons. These effects of Ex4 were not influenced by intracellular GDP-ß-S, indicating that GLP-1 signaling directly stimulates a population of axon terminals innervating the POMC neurons. The different Ex4 responsiveness of their mPSCs indicates the heterogeneity of the POMC neurons of the ARC. In summary, our data demonstrate that in addition to its direct excitatory effect on the POMC neurons, GLP-1 signaling also facilitates the presynaptic input of these cells by acting on presynaptically localized GLP-1R.


Assuntos
Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Exenatida/farmacologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Hipoglicemiantes/farmacologia , Neurônios/efeitos dos fármacos , Pró-Opiomelanocortina/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peptídeo 1 Semelhante ao Glucagon/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo
2.
Aging (Albany NY) ; 12(3): 2101-2122, 2020 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-32007953

RESUMO

As the elderly population grows, chronic metabolic dysfunction including obesity and diabetes are becoming increasingly common comorbidities. Hypothalamic inflammation through CNS resident microglia serves as a common pathway between developing obesity and developing systemic aging pathologies. Despite understanding aging as a life-long process involving interactions between individuals and their environment, limited studies address the dynamics of environment interactions with aging or aging therapeutics. We previously demonstrated environmental enrichment (EE) is an effective model for studying improved metabolic health and overall healthspan in mice, which acts through a brain-fat axis. Here we investigated the CSF1R inhibitor PLX5622 (PLX), which depletes microglia, and its effects on metabolic decline in aging in interaction with EE. PLX in combination with EE substantially improved metabolic outcomes in middle-aged female mice over PLX or EE alone. Chronic PLX treatment depleted 75% of microglia from the hypothalamus and reduced markers of inflammation without affecting brain-derived neurotrophic factor levels induced by EE. Adipose tissue remodeling and adipose tissue macrophage modulation were observed in response to CSF1R inhibition, which may contribute to the combined benefits seen in EE with PLX. Our study suggests benefits exist from combined drug and lifestyle interventions in aged animals.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Envelhecimento/metabolismo , Abrigo para Animais , Microglia/efeitos dos fármacos , Compostos Orgânicos/farmacologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Meio Social , Tecido Adiposo/metabolismo , Animais , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hormônio Liberador da Corticotropina/efeitos dos fármacos , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Teste de Tolerância a Glucose , Hormônio Liberador de Gonadotropina/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Inflamação/genética , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Neuropeptídeo Y/efeitos dos fármacos , Neuropeptídeo Y/genética , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transcriptoma/efeitos dos fármacos , Redução de Peso
3.
Endocr J ; 67(2): 177-184, 2020 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-31666445

RESUMO

Cushing's disease is primarily caused by autonomic hypersecretion of adrenocorticotropic hormone (ACTH) from a pituitary adenoma. In Cushing's disease, mutations in the ubiquitin-specific protease 8 (USP8) have been detected. These mutations are associated with hyperactivation of USP8 that prevent epidermal growth factor receptor (EGFR) degradation. This leads to increased EGFR stability and results in the maintenance of EGFR signaling in Cushing's disease. USP8 inhibitors can suppress the growth of various tumors. In this study, the effects of a potent USP8 inhibitor, DUBs-IN-2, on ACTH production and cell proliferation were examined in mouse corticotroph tumor (AtT-20) cells. Proopiomelanocortin (Pomc) mRNA levels and ACTH levels were decreased in AtT-20 cells by DUBs-IN-2. Further, cell proliferation was inhibited, and apoptosis was induced by DUBs-IN-2. Transcript levels of pituitary tumor-transforming gene 1 (Pttg1), a pituitary tumor growth marker, were increased; and transcript levels of stress response growth arrest and DNA damage-inducible 45 (Gadd45ß) and Cdk5 and ABL enzyme substrate 1 (Cables1) mRNA levels were increased in response to the drug. Gadd45ß or Cables1 knockdown partially inhibited the DUBs-IN-2-induced decrease in cell proliferation, but not Pomc mRNA levels. Both GADD45ß and CABLES1 may be responsible, at least in part, for the USP8-induced suppression of corticotroph tumor cell proliferation. USP-8 may be a new treatment target in Cushing's disease.


Assuntos
Adenoma Hipofisário Secretor de ACT/metabolismo , Adenoma/metabolismo , Hormônio Adrenocorticotrópico/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Complexos Endossomais de Distribuição Requeridos para Transporte/antagonistas & inibidores , Indenos/farmacologia , Pirazinas/farmacologia , Ubiquitina Tiolesterase/antagonistas & inibidores , Hormônio Adrenocorticotrópico/metabolismo , Animais , Antígenos de Diferenciação/efeitos dos fármacos , Antígenos de Diferenciação/genética , Linhagem Celular Tumoral , Quinase 5 Dependente de Ciclina/efeitos dos fármacos , Quinase 5 Dependente de Ciclina/genética , Ciclinas/efeitos dos fármacos , Ciclinas/genética , Endopeptidases , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , Hipersecreção Hipofisária de ACTH/metabolismo , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/genética , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Securina/efeitos dos fármacos , Securina/genética
4.
Brain Behav ; 9(9): e01340, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31392839

RESUMO

INTRODUCTION: Ginseng polysaccharide (GPS, same as Panax polysaccharide) is a kind of polysaccharide extracted from ginseng. It has been reported that GPS has the ability to activate innate immunity, regulates blood sugar balance, and improves antioxidant capacity, but the effect on feeding behavior and its mechanism remains unclear. METHOD: To investigate the possible effect of GPS on feeding behavior of animals, mice were supplied with GPS in water, and food intake, hedonic feeding behavior, anxiety-like behavior, expression of appetite-regulation peptides in the central nervous system and glucose-related hormone levels in the serum of mice were measured. RESULTS: Ginseng polysaccharide significantly increased the average daily food intake in mice and promoted hedonic eating behavior. Meanwhile, the levels of serum glucose and glucagon were significantly reduced by GPS, and GPS promoted hypothalamic neuropeptide Y expression, inhibited proopiomelanocortin (POMC) expression, and reduced dopamine D1 receptor (DRD1) levels in the midbrain. We also found that the anxiety level of mice was significantly lower after GPS intake. In conclusion, oral supplementation with GPS promoted food intake in mice, most likely through the regulation of circulating glucose levels.


Assuntos
Comportamento Alimentar/efeitos dos fármacos , Panax , Polissacarídeos/farmacologia , Animais , Ansiedade , Comportamento Animal/efeitos dos fármacos , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Suplementos Nutricionais , Proteínas da Membrana Plasmática de Transporte de Dopamina/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Ingestão de Alimentos/efeitos dos fármacos , Glucagon/efeitos dos fármacos , Glucagon/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Insulina/metabolismo , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Camundongos , Neuropeptídeo Y/efeitos dos fármacos , Neuropeptídeo Y/metabolismo , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores de Dopamina D1/efeitos dos fármacos , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/efeitos dos fármacos , Receptores de Dopamina D2/genética
5.
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
6.
J Clin Endocrinol Metab ; 102(8): 2825-2835, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28505327

RESUMO

Context: Remission failure following transsphenoidal surgery in Cushing disease (CD) from pituitary corticotroph tumors (CtTs) remains clinically challenging. Histone deacetylase inhibitors (HDACis) are antitumor drugs approved for clinical use, with the potential to affect adrenocorticotropin hormone (ACTH) hypersecretion by inhibiting pro-opiomelanocortin (POMC) transcription. Objective: Testing the efficacy of suberoylanilide hydroxamic acid (SAHA) on human and murine ACTH-secreting tumor (AtT-20) cells. Design: Cell viability, ACTH secretion (enzyme-linked immunosorbent assay), apoptosis, and gene expression profile were investigated on AtT-20 cells. In vivo efficacy was examined in an athymic nude mouse AtT-20 xenograft model. SAHA efficacy against human-derived corticotroph tumor (hCtT) (n = 8) was tested in vitro. Setting: National Institutes of Health. Intervention: SAHA (0.5 to 8 µM). Main Outcome Measures: AtT-20 and hCtT cell survival, in vitro/invivo ACTH measurements. Results: SAHA (1 µM) reduced AtT-20 viability to 75% at 24 hours, 43% at 48 hours (analysis of variance; P = 0.002). Apoptosis was confirmed with elevated BAX/Bcl2 ratio and FACS. Intriguingly, early (3-hour) significant decline (70%; P < 0.0001) of secreted ACTH and diminished POMC transcription was observed with SAHA (1 µM). Microarray analysis revealed a direct association between liver X receptor alpha (LXRα) and POMC expression. Accordingly, SAHA reduced LXRα in AtT-20 cells but not in normal murine corticotrophs. Xenografted nude-mice tumor involution (126 ± 33/160 ± 35 vs 337 ± 49 mm3; P = 0.0005) was observed with 5-day intraperitoneal SAHA, with reversal of elevated ACTH (P < 0.0001). SAHA did not affect serum ACTH in nontumor mice. Lastly, we confirmed that SAHA (1 µM/24 h) decreased hCtT survival (78.92%; P = 0.0007) and ACTH secretion (83.64%; P = 0.03). Conclusion: Our findings demonstrate SAHA's efficacy in reducing survival and ACTH secretion in AtT-20 and hCtT cells, providing a potential intervention for recurrent/unremitting CD.


Assuntos
Adenoma Hipofisário Secretor de ACT/tratamento farmacológico , Adenoma/tratamento farmacológico , Hormônio Adrenocorticotrópico/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Hipersecreção Hipofisária de ACTH/tratamento farmacológico , Adenoma Hipofisário Secretor de ACT/metabolismo , Adenoma/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Corticotrofos/citologia , Corticotrofos/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Técnicas In Vitro , Camundongos , Camundongos Nus , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/genética , Reação em Cadeia da Polimerase em Tempo Real , Vorinostat , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Anesthesiology ; 127(2): 347-354, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28542000

RESUMO

BACKGROUND: We compared the effects of etomidate and ketamine on the hypothalamic-pituitary-adrenal axis during sepsis. METHODS: Mice (n = 5/group) were injected intraperitoneally with lipopolysaccharide (10 mg/kg) and 6 h later randomized to receive ketamine (100 mg/kg), etomidate (30 mg/kg), or saline. At two time points (12 and 48 h), messenger RNA levels of hypothalamic corticotropin-releasing hormone, pituitary proopiomelanocortin, and four adrenal enzymes (P450 side-chain cleavage, 3ß-hydroxysteroid deshydrogenase, 21-hydroxylase, and 11ß-hydroxylase) were measured by in situ hybridization (results are presented as optical density), and plasma levels of corticosterone and adrenocorticotropin hormones were measured by enzyme-linked immunosorbent assay (mean ± SD). RESULTS: At 12 h, lipopolysaccharide induced an overexpression of corticotropin-releasing hormone (32 ± 5 vs. 18 ± 6, P < 0.01), proopiomelanocortin (21 ± 3 vs. 8 ± 0.9, P < 0.0001), P450 side-chain cleavage (32 ± 4 vs. 23 ± 10, P < 0.05), 21-hydroxylase (17 ± 5 vs. 12 ± 2, P < 0.05), and 11ß-hydroxylase (11 ± 4 vs. 6 ± 0.5, P = 0.001), and an elevation of corticosterone (642 ± 165 vs. 98.3 ± 63 ng/ml, P < 0.0001). Etomidate and ketamine reduced P450 side-chain cleavage (19 ± 7 and 19 ± 3 vs. 32 ± 4, P < 0.01), 21-hydroxylase (8 ± 0.8 and 8 ± 1 vs. 17 ± 5, P < 0.001), 11ß-hydroxylase (4 ± 0.5 and 7 ± 1 vs. 11 ± 4, P < 0.001 and P < 0.05), and corticosterone (413 ± 189 and 260 ± 161 vs. 642 ± 165 ng/ml, P < 0.05 and P < 0.01). Ketamine also inhibited adrenocorticotropin hormone production (2.5 ± 3.6 vs. 36 ± 15 pg/ml, P < 0.05). At 48 h, all four adrenal enzymes were down-regulated by lipopolysaccharide administration with corticosterone levels similar to the control group. Ketamine and etomidate did not modify corticosterone plasma levels. CONCLUSIONS: Our endotoxemic model induces an initial activation of the hypothalamic-pituitary-adrenal axis, followed by a secondary inhibition of adrenal steroidogenesis processes. Ketamine and etomidate inhibit the enzyme expression and activity of the adrenal gland at the early stage.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Endotoxemia , Etomidato/farmacologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Ketamina/farmacologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Analgésicos/farmacologia , Animais , Corticosterona/sangue , Hormônio Liberador da Corticotropina/sangue , Hormônio Liberador da Corticotropina/efeitos dos fármacos , Modelos Animais de Doenças , Etomidato/sangue , Hipnóticos e Sedativos/farmacologia , Sistema Hipotálamo-Hipofisário/fisiopatologia , Ketamina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistema Hipófise-Suprarrenal/fisiopatologia , Pró-Opiomelanocortina/sangue , Pró-Opiomelanocortina/efeitos dos fármacos , Esteroide 21-Hidroxilase/sangue , Esteroide 21-Hidroxilase/efeitos dos fármacos
8.
Diabetes ; 66(4): 920-934, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28073831

RESUMO

Effectors of the phosphoinositide 3-kinase (PI3K) signal transduction pathway contribute to the hypothalamic regulation of energy and glucose homeostasis in divergent ways. Here we show that central nervous system (CNS) action of the PI3K signaling intermediate atypical protein kinase C (aPKC) constrains food intake, weight gain, and glucose intolerance in both rats and mice. Pharmacological inhibition of CNS aPKC activity acutely increases food intake and worsens glucose tolerance in chow-fed rodents and causes excess weight gain during high-fat diet (HFD) feeding. Similarly, selective deletion of the aPKC isoform Pkc-λ in proopiomelanocortin (POMC) neurons disrupts leptin action, reduces melanocortin content in the paraventricular nucleus, and markedly increases susceptibility to obesity, glucose intolerance, and insulin resistance specifically in HFD-fed male mice. These data implicate aPKC as a novel regulator of energy and glucose homeostasis downstream of the leptin-PI3K pathway in POMC neurons.


Assuntos
Ingestão de Alimentos/genética , Intolerância à Glucose/genética , Glucose/metabolismo , Isoenzimas/genética , Neurônios/metabolismo , Obesidade/genética , Proteína Quinase C/genética , Aumento de Peso/genética , Animais , Dieta Hiperlipídica , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Intolerância à Glucose/metabolismo , Hipotálamo/metabolismo , Resistência à Insulina , Leptina/metabolismo , Masculino , Melanocortinas/metabolismo , Camundongos , Obesidade/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Ratos , Transdução de Sinais , Aumento de Peso/efeitos dos fármacos
9.
Genet Mol Res ; 15(1): 15017362, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26985919

RESUMO

The hypothalamus is an important component of the nervous system, and neuropeptide Y (NPY), proopiomelanocortin (POMC), and neuromedin U (NMU) are key players in physiological regulation. Puerarin is important for nerve regulation. We investigated the effect of puerarin on the expression of NMU, NPY, and POMC genes in the hypothalamus. The results showed that the puerarin low-dose group and the other groups were significantly different (P < 0.05). However, there was no significant difference in NMU, POMC, and NPY among the groups.


Assuntos
Hipotálamo/metabolismo , Isoflavonas/farmacologia , Neuropeptídeo Y/genética , Neuropeptídeos/genética , Pró-Opiomelanocortina/genética , Animais , Regulação da Expressão Gênica , Hipotálamo/efeitos dos fármacos , Neuropeptídeo Y/efeitos dos fármacos , Neuropeptídeos/efeitos dos fármacos , Pró-Opiomelanocortina/efeitos dos fármacos , Ratos
10.
Pharmacol Res ; 102: 235-44, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26523876

RESUMO

Protein tyrosine phosphatase 1b (Ptp1b), which represses leptin signaling, is a promising therapeutic target for obesity. Genome wide deletion of Ptp1b, increases leptin sensitivity, protects mice from obesity and diabetes, but alters cardiovascular function by increasing blood pressure (BP). Leptin-control of metabolism is centrally mediated and involves proopiomelanocortin (POMC) neurons. Whether these neurons contribute to leptin-mediated increases in BP remain unclear. We hypothesized that increasing leptin signaling in POMC neurons with Ptp1b deletion will sensitize the cardiovascular system to leptin and enhance neurogenic control of BP. We analyzed the cardiovascular phenotype of Ptp1b+/+ and POMC-Ptp1b-/- mice, at baseline and after 7 days of leptin infusion or sympatho-activation with phenylephrine. POMCPtp1b deletion did not alter baseline cardiovascular hemodynamics (BP, heart rate) but reduced BP response to ganglionic blockade and plasma catecholamine levels that suggests a decreased neurogenic control of BP. In contrast, POMC-Ptp1b deletion increased vascular adrenergic reactivity and aortic α-adrenergic receptors expression. Chronic leptin treatment reduced vascular adrenergic reactivity and blunted diastolic and mean BP increases in POMC-Ptp1b-/- mice only. Similarly POMC-Ptp1b-/- mice exhibited a blunted increased in diastolic and mean BP accompanied by a gradual reduction in adrenergic reactivity in response to chronic vascular sympatho-activation with phenylephrine. Together these data rule out our hypothesis but suggest that deletion of Ptp1b in POMC neurons protects from leptin- and sympatho-mediated increases in BP. Vascular adrenergic desensitization appears as a protective mechanism against hypertension, and POMC-Ptp1b as a key therapeutic target for the treatment of metabolic and cardiovascular dysfunctions associated with obesity.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Hipertensão/sangue , Hipertensão/metabolismo , Leptina/farmacologia , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Animais , Metabolismo Energético/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Obesidade/metabolismo , Fenilefrina/farmacologia , Pró-Opiomelanocortina/efeitos dos fármacos , Receptores Adrenérgicos alfa/metabolismo
11.
Pituitary ; 18(4): 542-53, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25280813

RESUMO

PURPOSE: Cushing's disease is primarily caused by adrenocorticotropic hormone (ACTH)-producing pituitary adenomas. If excision of the tumor from the pituitary, which is the primary treatment for Cushing's disease, is unsuccessful, further medical therapy is needed to treat the resultant hypercortisolism. Some of the drugs used to treat this condition have shown potential therapeutic benefits, but a more effective treatment should be explored for the treatment of Cushing's disease. In the present study, we determined the effect of heat shock protein 90 inhibitors on ACTH production and cell proliferation of AtT-20 corticotroph tumor cells. METHODS: AtT-20 pituitary corticotroph tumor cells were cultured. The expression levels of mouse proopiomelanocortin (POMC) and pituitary tumor transforming gene 1 (PTTG1) mRNA were evaluated using quantitative real-time PCR. Cellular DNA content was analyzed with fluorescence-activated cell sorting (FACS) analysis. The protein levels were determined by Western blot analysis. RESULTS: Both 17-allylamino-17-demethoxygeldanamycin and CCT018159 decreased POMC mRNA levels in AtT-20 cells and ACTH levels in the culture medium of these cells, suggesting that both drugs suppress ACTH synthesis and secretion in corticotroph tumor cells. Both drugs also decreased cell proliferation and induced apoptosis. FACS analyses revealed that both agents increased the percentage of AtT-20 cells in the G2/M phase. These drugs decreased cell proliferation, presumably due to the induction of cell death and arrest of the cell cycle in AtT-20 cells. Tumor weight in mice xenografted with AtT-20 cells and treated with CCT018159 was lower than in AtT-20-xenografted control mice. CCT018159 also decreased plasma ACTH levels, and POMC and PTTG1 mRNA levels in the tumor cells. CONCLUSIONS: CCT018159 inhibits ACTH production and corticotroph tumor cell proliferation in vitro and in vivo.


Assuntos
Adenoma Hipofisário Secretor de ACT/genética , Adenoma/genética , Hormônio Adrenocorticotrópico/efeitos dos fármacos , Benzoquinonas/farmacologia , Proliferação de Células/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Lactamas Macrocíclicas/farmacologia , RNA Mensageiro/efeitos dos fármacos , Adenoma Hipofisário Secretor de ACT/metabolismo , Adenoma Hipofisário Secretor de ACT/patologia , Adenoma/metabolismo , Adenoma/patologia , Hormônio Adrenocorticotrópico/metabolismo , Animais , Linhagem Celular Tumoral , Compostos Heterocíclicos com 2 Anéis/farmacologia , Camundongos , Transplante de Neoplasias , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/genética , Pirazóis/farmacologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Securina/efeitos dos fármacos , Securina/genética , Carga Tumoral
12.
Endocrinology ; 154(4): 1465-75, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23493373

RESUMO

The endocrine disrupting compound bisphenol-A (BPA) has been reported to act as an obesogen in rodents exposed perinatally. In this study, we investigated the effects of early-life BPA exposure on adult metabolic phenotype and hypothalamic energy balance circuitry. Pregnant and lactating CD-1 dams were exposed, via specially prepared diets, to 2 environmentally relevant doses of BPA. Dams consumed an average of 0.19 and 3.49 µg/kg per day of BPA in the low and high BPA treatments prenatally and an average of 0.36 and 7.2 µg/kg per day of BPA postnatally. Offspring were weaned initially onto a normal (AIN93G) diet, then as adults exposed to either a normal or high-fat diet (HFD). Males exposed to the high dose of BPA showed impaired glucose tolerance on both diets. They also showed reduced proopiomelanocortin fiber innervation into the paraventricular nucleus of the hypothalamus, and when exposed to HFD, they demonstrated increased neuropeptide Y and Agouti-related peptide expression in the arcuate nucleus (ARC). Females exposed to the high BPA dose were heavier, ate more, and had increased adiposity and leptin concentrations with reduced proopiomelanocortin mRNA expression in the ARC when consuming a HFD. BPA-exposed females showed ARC estrogen receptor α expression patterns similar to those seen in males, suggesting a masculinizing effect of BPA. These results demonstrate that early-life exposure to the obesogen BPA leads to sexually dimorphic alterations in the structure of hypothalamic energy balance circuitry, leading to increased vulnerability for developing diet-induced obesity and metabolic impairments, such as glucose intolerance.


Assuntos
Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Compostos Benzidrílicos/efeitos adversos , Dietilestilbestrol/efeitos adversos , Estrogênios não Esteroides/efeitos adversos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Fenóis/efeitos adversos , Aumento de Peso/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Dieta Hiperlipídica , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Receptor alfa de Estrogênio/metabolismo , Comportamento Alimentar/efeitos dos fármacos , Feminino , Teste de Tolerância a Glucose , Masculino , Camundongos , Fibras Nervosas/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/metabolismo , Fatores Sexuais
13.
J Neurophysiol ; 106(3): 1191-202, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21653710

RESUMO

Two of the biggest health problems facing us today are addiction to nicotine and the increased prevalence of obesity. Interestingly, nicotine attenuates obesity, but the underlying mechanism is not clear. Here we address the hypothesis that if weight-reducing actions of nicotine are mediated by anorexigenic proopiomelanocortin (POMC) neurons of the hypothalamic arcuate nucleus, nicotine should excite these cells. Nicotine at concentrations similar to those found in smokers, 100-1,000 nM, excited POMC cells by mechanisms based on increased spike frequency, depolarization of membrane potential, and opening of ion channels. This was mediated by activation of both α7 and α4ß2 nicotinic receptors; by itself, this nicotine-mediated excitation could explain weight loss caused by nicotine. However, in control experiments nicotine also excited the orexigenic arcuate nucleus neuropeptide Y (NPY) cells. Nicotine exerted similar actions on POMC and NPY cells, with a slightly greater depolarizing action on POMC cells. Immunocytochemistry revealed cholinergic axons terminating on both cell types. Nicotine actions were direct in both cell types, with nicotine depolarizing the membrane potentials and reducing input resistance. We found no differences in the relative desensitization to nicotine between POMC and NPY neurons. Nicotine inhibited excitatory synaptic activity recorded in NPY, but not POMC, cells. Nicotine also excited hypocretin/orexin neurons that enhance cognitive arousal, but the responses were smaller than in NPY or POMC cells. Together, these results indicate that nicotine has a number of similar actions, but also a few different actions, on POMC and NPY neurons that could contribute to the weight loss associated with smoking.


Assuntos
Depressores do Apetite/farmacologia , Núcleo Arqueado do Hipotálamo/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neuropeptídeo Y/fisiologia , Neuropeptídeos/fisiologia , Nicotina/farmacologia , Pró-Opiomelanocortina/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Orexinas , Pró-Opiomelanocortina/efeitos dos fármacos
14.
Diabetes ; 59(10): 2418-25, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20682682

RESUMO

OBJECTIVE: Olanzapine (OLZ) is an atypical antipsychotic whose clinical efficacy is hampered by side effects including weight gain and diabetes. Recent evidence shows that OLZ alters insulin sensitivity independent of changes in body weight and composition. The present study addresses whether OLZ-induced insulin resistance is driven by its central actions. RESEARCH DESIGN AND METHODS: Sprague-Dawley rats received an intravenous (OLZ-IV group) or intracerebroventricular (OLZ-ICV group) infusion of OLZ or vehicle. Glucose kinetics were assessed before (basal period) and during euglycemic-hyperinsulinemic clamp studies. RESULTS: OLZ-IV caused a transient increase in glycemia and a higher rate of glucose appearance (R(a)) in the basal period. During the hyperinsulinemic clamp, the glucose infusion rate (GIR) required to maintain euglycemia and the rate of glucose utilization (R(d)) were decreased in OLZ-IV, whereas endogenous glucose production (EGP) rate was increased compared with vehicle-IV. Consistent with an elevation in EGP, the OLZ-IV group had higher hepatic mRNA levels for the enzymes glucose-6-phosphatase and phosphoenolpyruvate carboxykinase. Phosphorylation of hypothalamic AMP-activated protein kinase (AMPK) was increased in OLZ-IV rats compared with controls. Similarly, an intracerebroventricular infusion of OLZ resulted in a transient increase in glycemia as well as a higher R(a) in the basal period. During the hyperinsulinemic period, OLZ-ICV caused a decreased GIR, an increased EGP, but no change in R(d). Furthermore, OLZ-ICV rats had increased hepatic gluconeogenic enzymes and elevated hypothalamic neuropeptide-Y and agouti-related protein mRNA levels. CONCLUSIONS: Acute central nervous system exposure to OLZ induces hypothalamic AMPK and hepatic insulin resistance, pointing to a hypothalamic site of action for the metabolic dysregulation of atypical antipsychotics.


Assuntos
Antipsicóticos/farmacologia , Benzodiazepinas/farmacologia , Resistência à Insulina/fisiologia , Fígado/fisiologia , Animais , Antipsicóticos/administração & dosagem , Benzodiazepinas/administração & dosagem , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Artérias Carótidas/fisiopatologia , Primers do DNA , Técnica Clamp de Glucose , Glucose-6-Fosfatase/efeitos dos fármacos , Glucose-6-Fosfatase/genética , Hiperinsulinismo/fisiopatologia , Infusões Intravenosas , Injeções Intraventriculares , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Veias Jugulares/fisiopatologia , Cinética , Fígado/efeitos dos fármacos , Masculino , Neuropeptídeos/efeitos dos fármacos , Neuropeptídeos/genética , Olanzapina , Orexinas , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/genética , Ratos , Ratos Sprague-Dawley , Tubulina (Proteína)/efeitos dos fármacos , Tubulina (Proteína)/genética
15.
Toxicol Appl Pharmacol ; 233(2): 314-22, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18824019

RESUMO

The mechanism by which a single administration of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) reduces food and water intake is unclear. We examined whether such a food and water intake-reducing single administration of TCDD induced changes in corticotropin-releasing factor (CRF), arginine vasopressin (AVP), and proopiomelanocortin (POMC) expression in rat brain. To observe time-dependent changes in these neuropeptides, male Sprague-Dawley rats were given TCDD (50 microg/kg) and terminated 1, 2, 4, or 7 days later. In addition, to observe dose-dependent changes in feeding and neuropeptides, rats were also given a range of TCDD doses (12.5, 25, or 50 microg/kg) and terminated 14 days later. TCDD suppressed food and water intake over 14 days in a dose-dependent manner. TCDD treatment also increased CRF and POMC mRNA levels in the hypothalamic paraventricular nucleus (PVN) and arcuate nucleus, respectively, in a dose- and time-dependent manner. These increases were related to decreased food intake following TCDD administration. TCDD treatment increased AVP and CRF mRNA levels in the PVN, and these increases were related to decreased water intake. Interestingly, the increases in CRF, AVP and POMC expression were observed 7 to 14 days after TCDD administration. These results suggest that a single administration of TCDD induced long-lasting increases in CRF, AVP, and POMC mRNA levels in the hypothalamus and that these changes are related to reduced food and water intake 7 to 14 days after TCDD administration.


Assuntos
Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Animais , Arginina Vasopressina/efeitos dos fármacos , Arginina Vasopressina/metabolismo , Hormônio Liberador da Corticotropina/efeitos dos fármacos , Hormônio Liberador da Corticotropina/metabolismo , Relação Dose-Resposta a Droga , Poluentes Ambientais/administração & dosagem , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Dibenzodioxinas Policloradas/administração & dosagem , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
16.
Lasers Surg Med ; 39(10): 797-802, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18081143

RESUMO

BACKGROUND: Low-level laser therapy (LLLT) has been reported to relieve pain with minimal side effects. Recent studies have demonstrated that opioid-containing immune cells migrate to inflamed sites and release beta-endorphins to inhibit pain as a mode of peripheral endogenous opioid analgesia. The present study investigates whether LLLT may enhance peripheral endogenous opioid analgesia. METHODS: The effect of LLLT on opioid analgesia and production was evaluated in vivo in a rat model of inflammation as well as in vitro in Jurkat cells, a human T-cell leukemia cell line. mRNA expression of the beta-endorphin precursors proopiomelanocortin and corticotrophin releasing factor was assessed by reverse transcription polymerase chain reaction. RESULTS: LLLT produced an analgesic effect in inflamed peripheral tissue which was transiently antagonized by naloxone. Beta-endorphin precursor mRNA expression increased with LLLT, both in vivo and in vitro. CONCLUSION: This study demonstrates that LLLT produces analgesic effects in a rat model of peripheral inflammation. We further revealed an additional mechanism of LLLT-mediated analgesia via enhancement of peripheral endogenous opioids. These findings suggest that LLLT induces analgesia in rats by enhancing peripheral endogenous opioid production in addition to previously reported mechanisms.


Assuntos
Hormônio Adrenocorticotrópico/metabolismo , Hormônios/metabolismo , Terapia com Luz de Baixa Intensidade , Pró-Opiomelanocortina/metabolismo , Adjuvantes Imunológicos/farmacologia , Hormônio Adrenocorticotrópico/efeitos dos fármacos , Animais , Adjuvante de Freund/farmacologia , Membro Posterior/patologia , Humanos , Imuno-Histoquímica , Células Jurkat , Masculino , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Pró-Opiomelanocortina/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Coloração e Rotulagem
17.
Neurosci Lett ; 423(2): 153-7, 2007 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-17693022

RESUMO

Although the orexigenic action of peptide hormones such as ghrelin and growth hormone releasing peptide is different between chickens and mammals, the anorexigenic action of peptide hormones is similar in both species. For example, central administration of peptide hormones such as leptin, cholecystokinin or glucagon has been shown to suppress food intake behavior in chickens and mammals. Central administration of insulin suppresses food intake in mammals. However, the anorexigenic action of insulin in chickens has not yet been identified. In the present study, we investigated the effects of central administration of insulin on food intake in chicks. Intracerebroventricular administration of insulin in chicks significantly suppressed food intake. Central administration of insulin significantly upregulated mRNA levels of proopiomelanocortin (POMC), cocaine- and amphetamine-regulated transcript (CART) and corticotropin-releasing factor (CRF), but did not influence mRNA levels of neuropeptide Y and agouti-related protein in the hypothalamus. These results suggest that alpha-melanocyte stimulating hormone (alpha-MSH, an anorexigenic peptide from the post-translational cleavage of POMC), CART and CRF are involved in the anorexigenic action of insulin in chicks. Furthermore, central administration of alpha-MSH or CART significantly suppressed food intake. In addition, alpha-MSH significantly upregulated CRF mRNA expression, suggesting that the anorexigenic action of alpha-MSH is mediated by CRF. Our findings demonstrate that insulin functions in chicks as an appetite-suppressive peptide in the central nervous system and suggest that this anorexigenic action is mediated by CART, alpha-MSH and CRF.


Assuntos
Regulação do Apetite/fisiologia , Encéfalo/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Insulina/administração & dosagem , Animais , Animais Recém-Nascidos , Encéfalo/fisiologia , Galinhas , Hormônio Liberador da Corticotropina/biossíntese , Hormônio Liberador da Corticotropina/efeitos dos fármacos , Relação Dose-Resposta a Droga , Injeções Intraventriculares , Masculino , Hormônios Estimuladores de Melanócitos/biossíntese , Hormônios Estimuladores de Melanócitos/efeitos dos fármacos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/efeitos dos fármacos , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/efeitos dos fármacos , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Phytomedicine ; 13(9-10): 658-67, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16516452

RESUMO

Koso-san (Xiang-Su-San in Chinese), a Kampo (Japanese herbal) medicine, is used clinically in East Asia for the treatment of depression-like symptoms associated with the initial stage of the common cold, allergic urticaria due to food ingestion, irritable bowel syndrome, chronic fatigue syndrome, insomnia, and autonomic imbalance. However, the antidepressant-like activity of Koso-san has never been evaluated scientifically. In this study, ddY mice subjected to a combination of forced swimming and chronic mild stresses were termed depression-like model mice. The degree of the depression-like state was measured by the animal's duration of immobility using the forced swimming test (FST). Oral administration of Koso-san (1.0 g/kg/body wt./day, 9 days) significantly shortened the duration of immobility of the depression-like model mice in the FST; however, locomotor activity was not affected. Hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis plays an important role in the pathophysiology of depression. Levels of corticotropin-releasing hormone mRNA expression in the hypothalamus and proopiomelanocortin mRNA expression in the pituitary were significantly increased, and glucocorticoid receptor protein expression in the hypothalamus paraventricular nucleus was downregulated in the depression-like model mice. However, Koso-san ameliorated these alterations to the normal conditions. The results of this study suggest that Koso-san shows the antidepressant-like effect through suppressing the hyperactivity of the HPA axis in depression-like model mice.


Assuntos
Antidepressivos/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Animais , Antidepressivos/análise , Corticosterona/sangue , Hormônio Liberador da Corticotropina/efeitos dos fármacos , Diazepam/farmacologia , Medicamentos de Ervas Chinesas/química , Expressão Gênica/efeitos dos fármacos , Hipnóticos e Sedativos/farmacologia , Hipotálamo/efeitos dos fármacos , Resposta de Imobilidade Tônica/efeitos dos fármacos , Masculino , Medicina Kampo , Camundongos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Pró-Opiomelanocortina/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , Receptores de Glucocorticoides/efeitos dos fármacos , Natação
19.
J Neuroendocrinol ; 18(2): 129-38, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16420282

RESUMO

This study examined the effects of the glucocorticoid receptor (GR) agonist RU28362 on stress-induced gene expression in the pituitary of rats to investigate mechanisms of glucocorticoid negative feedback in vivo. In an initial experiment, acute restraint stress produced rapid (within 15 min) induction of c-fos mRNA, zif268 mRNA and pro-opiomelanocortin (POMC) hnRNA within the anterior and intermediate/posterior pituitary as determined by quantitative real-time polymerase chain reaction. Treatment with RU28362 (150 microg/kg, i.p.) 60 min before restraint inhibited adrenocorticotrophic hormone (ACTH) and corticosterone secretion and selectively suppressed the stress-induced increase in POMC hnRNA in the anterior pituitary gland. The failure of RU28362 to surpress the stress-induced rise in c-fos and expression of zif268 mRNA suggests that the central release of ACTH secretagogues was not affected at this time point by treatment with the GR agonist. Rather, the inhibition of ACTH release appeared to be due to a direct effect of RU28362 within the pituitary. A follow-up time-course study varied the interval (10, 60 or 180 min) between RU28362 pretreatment and the onset of restraint. The stress-induced increase in POMC hnRNA was completely blunted by RU28362 treatment within 10 min of treatment, although the stress induced hormone secretion, c-fos mRNA and zif268 mRNA were unaffected. The rapid inhibition of the stress-induced rise in POMC hnRNA in the anterior pituitary appears to reflect direct, GR-mediated suppression of POMC gene expression. RU28362 pretreatment 180 min before restraint onset was sufficient to suppress the stress-induced expression in the anterior pituitary gland of all three genes examined. Thus, the delayed negative feedback effects on hypothalamic-pituitary-adrenal axis activity that emerged after 180 min after glucocorticoid treatment were not evident at 60 min. Taken together, the data suggest that the inhibition of the stress-induced release of ACTH apparent within the first hour of glucocorticoid exposure is effected at the level of the pituitary gland. The delayed glucocorticoid effects evident 180 min after RU28362 treatment may include glucocorticoid actions in the brain and additional actions within the pituitary.


Assuntos
Androstanóis/farmacologia , Hipófise/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptores de Glucocorticoides/agonistas , Estresse Psicológico/metabolismo , Hormônio Adrenocorticotrópico/sangue , Hormônio Adrenocorticotrópico/efeitos dos fármacos , Animais , Corticosterona/sangue , Proteína 1 de Resposta de Crescimento Precoce/efeitos dos fármacos , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Retroalimentação Fisiológica , Regulação da Expressão Gênica/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Hipófise/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/genética , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Nuclear Heterogêneo/análise , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/metabolismo , Estatísticas não Paramétricas , Estresse Psicológico/genética , Fatores de Tempo
20.
Neuroscience ; 134(4): 1391-7, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16039786

RESUMO

In humans, stress is recognized as a major factor contributing to relapse to drug abuse in abstinent individuals; drugs of abuse themselves or withdrawal from such drugs act as stressors. In the animals, evidence suggests that centrally released arginine vasopressin in both amygdala and hypothalamus plays an important role in stress-related anxiogenic behaviors. The stress responsive hypothalamic-pituitary-adrenal axis is under tonic inhibition via endogenous opioids, and cocaine withdrawal stimulates hypothalamic-pituitary-adrenal activity. The present studies were undertaken to determine whether: (1) 14-day (chronic) "binge" pattern cocaine administration (45 mg/kg/day) or its withdrawal for 3 h (acute), 1 day (subacute) or 10 days (chronic) alters arginine vasopressin mRNA levels in amygdala or hypothalamus; (2) the opioid receptor antagonist naloxone (1mg/kg) alters arginine vasopressin mRNA or hypothalamic-pituitary-adrenal hormonal responses in acute cocaine withdrawal; and (3) there are associated changes of mu opioid receptor or proopiomelanocortin mRNA levels. In amygdala, arginine vasopressin mRNA levels were unchanged after chronic "binge" cocaine, but were increased during acute cocaine withdrawal. Naloxone completely blocked this increase. Neither chronic cocaine nor its acute withdrawal altered amygdalar mu opioid receptor mRNA levels. The increase in amygdalar arginine vasopressin mRNA levels was still observed after subacute withdrawal, but not after chronic withdrawal. Although hypothalamic-pituitary-adrenal tolerance developed with chronic "binge" cocaine, there were modestly elevated plasma adrenocorticotropin hormone levels during acute withdrawal. While naloxone produced modest adrenocorticotropin hormone elevations in cocaine-naïve rats, naloxone failed to elicit an adrenocorticotropin hormone response in cocaine-withdrawn rats. In hypothalamus, neither chronic cocaine nor acute withdrawal altered arginine vasopressin, proopiomelanocortin or mu opioid receptor mRNA levels. These results show that: (1) opioid receptors mediate increased amygdalar arginine vasopressin gene expression during acute cocaine withdrawal, and (2) cocaine withdrawal renders the hypothalamic-pituitary-adrenal axis insensitive to naloxone. Our findings suggest a potential role for amygdalar arginine vasopressin in the aversive consequences of early cocaine withdrawal.


Assuntos
Tonsila do Cerebelo/metabolismo , Arginina Vasopressina/efeitos dos fármacos , Arginina Vasopressina/metabolismo , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Animais , Arginina Vasopressina/genética , Expressão Gênica , Sistema Hipotálamo-Hipofisário/fisiologia , Hipotálamo/metabolismo , Masculino , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Sistema Hipófise-Suprarrenal/fisiologia , Pró-Opiomelanocortina/efeitos dos fármacos , Pró-Opiomelanocortina/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Endogâmicos F344 , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...