Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 299
Filtrar
1.
Horm Metab Res ; 45(13): 975-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24048879

RESUMO

Hunger and satiety are regulated in a complex fashion by a few food intake stimulatory (orexigenic) and a multitude of inhibitory (anorexigenic) factors produced in the periphery (mainly in the gastrointestinal tract) or directly in the brain. Within the brain, the hypothalamus plays a pivotal role as a production site of food intake regulatory factors. Importantly, this site integrates peripheral and central signaling factors to orchestrate food intake and in the long term body weight. Our knowledge on these regulatory pathways is not static but rather rapidly changing as new factors as well as up- and downstream signaling pathways of already known transmitters are uncovered. Hypothalamic nucleobindin2 (NUCB2), the precursor of nesfatin-1, was first described in 2006 and nesfatin-1 found to be a novel anorexigenic modulator of food intake and body weight. The initial report stimulated several groups to investigate the biological actions of nesfatin-1 and subsequent studies delineated the underlying brain mechanisms involved in its food reducing effect. Of interest was the demonstration that NUCB2 also exerts its anorexigenic action in the paraventricular nucleus of the hypothalamus and is regulated at this site by changes in metabolic status with a diurnal rhythm inversely related to that of feeding in rats. The present review describes the current state-of-knowledge on central nesfatin-1's effects on food intake and body weight and highlights important missing links regarding cellular signaling mechanisms involved in nesfatin-1's action.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação a DNA/metabolismo , Metabolismo Energético/fisiologia , Homeostase/fisiologia , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Peso Corporal/fisiologia , Ingestão de Alimentos/fisiologia , Humanos , Nucleobindinas , Ratos
2.
Gut ; 59(6): 744-51, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20551459

RESUMO

OBJECTIVE: Chronic psychological stress is associated with development of intestinal barrier dysfunction and impairs host defence mechanisms. The intestinal epithelium, consisting of enterocytes, endocrine cells, goblet cells and Paneth cells, is an important component of this barrier. In the present study, the impact of maternal deprivation (MD) on secretory lineages of duodenal epithelium and the involvement of the peripheral corticotropin-releasing factor (CRF) pathway were investigated. METHODS: Rat pups were deprived of their dam for 3 h/day (days 5-20). Non-deprived pups served as controls. On days 8, 13, 20, 24, 34, 44 and 84, duodenal tissues were collected for quantitative real-time PCR and immunohistochemistry studies. RESULTS: MD induced a sustained decrease in the number of Paneth and goblet cells but hyperplasia of endocrine cells. These alterations were associated with a duodenal increase of CRF, urocortin 2 and CRF receptor subtype 2 (CRFR(2)) mRNA, whereas CRFR(1) expression was decreased. The effects of MD on intestinal epithelium were inhibited by the CRFR(1)/R(2) antagonist astressin injected daily before MD. Studies using specific receptor antagonists in rats subjected to MD revealed that CRFR(1) was involved in the hyperplasia of endocrine cells and CRFR(2) in the depletion of Paneth cells. Conversely, daily injection of CRF and of the CRFR(2) agonist urocortin 2 in control rats resulted in changes in epithelial differentiation similar to MD. CONCLUSIONS: The activation of CRFR(1) and CRFR(2) induced by MD markedly altered the quantitative distribution of secretory cells of the intestinal epithelium. These alterations, in particular the depletion of Paneth and goblet cells, may create conditions leading to the development of an epithelial barrier defect.


Assuntos
Hormônio Liberador da Corticotropina/fisiologia , Duodeno/patologia , Mucosa Intestinal/patologia , Privação Materna , Estresse Psicológico/patologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Hormônio Liberador da Corticotropina/antagonistas & inibidores , Hormônio Liberador da Corticotropina/farmacologia , Duodeno/efeitos dos fármacos , Duodeno/metabolismo , Duodeno/fisiopatologia , Células Enteroendócrinas/patologia , Células Caliciformes/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiopatologia , Celulas de Paneth/patologia , Fragmentos de Peptídeos/farmacologia , Ratos , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Fatores de Transcrição/metabolismo , Urocortinas/farmacologia
3.
Science ; 222(4626): 935-7, 1983 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-6415815

RESUMO

Intracisternal injection of ovine corticotropin-releasing factor (CRF) into the pylorus-ligated rat or the rat with gastric fistula resulted in a dose-dependent inhibition of gastric secretion stimulated with pentagastrin or thyrotropin-releasing hormone. When injected into the lateral hypothalamus--but not when injected into the cerebral cortex--CRF suppressed pentagastrin-stimulated acid secretion. The inhibitory effect of CRF was blocked by vagotomy and adrenalectomy but not by hypophysectomy or naloxone treatment. These results indicate that CRF acts within the brain to inhibit gastric acid secretion through vagal and adrenal mechanisms and not through hypophysiotropic effects.


Assuntos
Encéfalo/efeitos dos fármacos , Hormônio Liberador da Corticotropina/farmacologia , Ácido Gástrico/metabolismo , Adrenalectomia , Animais , Córtex Cerebral/efeitos dos fármacos , Hormônio Liberador da Corticotropina/administração & dosagem , Relação Dose-Resposta a Droga , Hipofisectomia , Hipotálamo/efeitos dos fármacos , Masculino , Pentagastrina/antagonistas & inibidores , Ratos , Ratos Endogâmicos , Hormônio Liberador de Tireotropina/antagonistas & inibidores , Vagotomia
4.
Endocrinology ; 148(12): 6115-23, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17932219

RESUMO

Interactions between gastrointestinal signals are a part of integrated systems regulating food intake (FI). We investigated whether cholecystokinin (CCK)-8 and urocortin systems potentiate each other to inhibit FI and gastric emptying (GE) in fasted mice. Urocortin 1 and urocortin 2 (1 microg/kg) were injected ip alone or with CCK (3 microg/kg) in lean, diet-induced obese (DIO) or corticotropin-releasing factor receptor-2 (CRF(2))-deficient mice. Gastric vagal afferent activity was recorded from a rat stomach-vagus in vitro preparation. When injected separately, urocortin 1, urocortin 2, or CCK did not modify the 4-h cumulative FI in lean mice. However, CCK plus urocortin 1 or CCK plus urocortin 2 decreased significantly the 4-h FI by 39 and 27%, respectively, compared with the vehicle + vehicle group in lean mice but not in DIO mice. Likewise, CCK-urocortin-1 delayed GE in lean but not DIO mice, whereas either peptide injected alone at the same dose had no effect. CCK-urocortin 2 suppression of FI was observed in wild-type but not CRF(2)-deficient mice. Gastric vagal afferent activity was increased by intragastric artery injection of urocortin 2 after CCK at a subthreshold dose, and the response was reversed by devazepide. These data establish a peripheral synergistic interaction between CCK and urocortin 1 or urocortin 2 to suppress FI and GE through CRF(2) receptor in lean mice that may involve CCK modulation of gastric vagal afferent responsiveness to urocortin 2. Such synergy is lost in DIO mice, suggesting a resistance to the satiety signaling that may contribute to maintain obesity.


Assuntos
Colecistocinina/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Saciação/efeitos dos fármacos , Urocortinas/farmacologia , Animais , Colecistocinina/administração & dosagem , Sinergismo Farmacológico , Esvaziamento Gástrico/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Modelos Biológicos , Obesidade/fisiopatologia , Fragmentos de Peptídeos/administração & dosagem , Ratos , Receptores de Hormônio Liberador da Corticotropina/genética , Transdução de Sinais/efeitos dos fármacos , Estômago/efeitos dos fármacos , Estômago/inervação , Estômago/fisiologia , Urocortinas/administração & dosagem , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiologia
5.
Regul Pept ; 141(1-3): 1-7, 2007 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-17321609

RESUMO

Obestatin is a newly identified ghrelin-associated peptide (GAP) that is derived from post-translational processing of the prepro-ghrelin gene. Obestatin has been reported initially to be the endogenous ligand for the orphan receptor G protein-coupled receptor 39 (GPR39), and to reduce refeeding- and ghrelin-stimulated food intake and gastric transit in fasted mice, and body weight gain upon chronic peripheral injection. However, recent reports indicate that obestatin is unlikely to be the endogenous ligand for GPR39 based on the lack of specific binding on GRP39 receptor expressing cells and the absence of signal transduction pathway activation. In addition, a number of studies provided convergent evidence that ghrelin injected intracerebroventricularly or peripherally did not influence food intake, body weight gain, gastric transit, gastrointestinal motility, and gastric vagal afferent activity, as well as pituitary hormone secretions, in rats or mice. Similarly, obestatin did not alter ghrelin-induced stimulation of food intake or gastric transit. Therefore, the present state-of-knowledge on obestatin and GPR39 is leaving many unanswered questions that deserve further consideration. Those relate not only to redefining the biological action of obestatin that should be renamed GAP, but also the identification of the native ligand for GPR39.


Assuntos
Ingestão de Alimentos , Hormônios Peptídicos/química , Hormônios Peptídicos/fisiologia , Peptídeos/química , Terminologia como Assunto , Animais , Grelina , Humanos , Hormônios Peptídicos/genética , Processamento de Proteína Pós-Traducional , Receptores Acoplados a Proteínas G/metabolismo
6.
Neurogastroenterol Motil ; 19(3): 161-5, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17300284

RESUMO

Ghrelin is a gut peptide well established to induce prokinetic and appetite stimulatory actions. Obestatin is a novel 23-amino acid peptide derived from the processing of the ghrelin gene. The peptide name was in keeping with its initially reported actions to suppress food intake and digestive motility and to antagonize ghrelin's stimulatory effect through interaction with the orphan GPR-39 receptor. However, subsequently, these findings have been questioned because obestatin actions to reduce food intake and to inhibit gastrointestinal (GI) motility in vivo and in vitro have not been reproduced by several groups. Furthermore, while GPR-39 appears to be involved in gut motor functions, convergent reports showed that obestatin is not the cognate ligand for this receptor. In light of recent controversy over the effects of obestatin, the present findings from De Smet et al. provides additional evidence that obestatin does not influence food intake and GI motility in vivo and in vitro. Taken together, existing reports curtail the initial promise that obestatin is a new regulator of appetite and digestive motility. Therefore, it is proposed to rename obestatin as ghrelin-associated peptide.


Assuntos
Regulação do Apetite/fisiologia , Motilidade Gastrointestinal/fisiologia , Hormônios Peptídicos/fisiologia , Animais , Grelina , Humanos , Hormônios Peptídicos/genética , Hormônios Peptídicos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
7.
Neurogastroenterol Motil ; 19(11): 923-36, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17973638

RESUMO

Intraperitoneal (i.p.) corticotropin releasing factor (CRF) induced a CRF(1) receptor-dependent stimulation of myenteric neurons and motility in the rat proximal colon. We characterize the colonic enteric nervous system response to CRF in conscious rats. Laser capture microdissection combined with reverse transcriptase polymerase chain reaction (RT-PCR) and immunohistochemistry in longitudinal muscle myenteric plexus whole-mount colonic preparations revealed CRF(1) receptor expression in myenteric neurons. CRF (i.p., 10 microg kg(-1)) induced Fos immunoreactivity (IR) (cells per ganglion) selectively in myenteric plexus of proximal (18.3 +/- 2.4 vs vehicle: 0.0 +/- 0.0) and distal colon (16.8 +/- 1.2 vs vehicle: 0.0 +/- 0.0), but not in that of gastric corpus, antrum, duodenum, jejunum and ileum. The selective CRF(1) agonist, stressin(1)-A (i.p., 10 microg kg(-1)) also induced Fos IR in myenteric but not in submucosal plexus of the proximal and distal colon. Fos IR induced by CRF was located in 55 +/- 1.9% and 53 +/- 5.1% of CRF(1) receptor-IR myenteric neurons and in 44 +/- 2.8% and 40 +/- 3.9% of cholinergic neurons with Dogiel type I morphology, and in 20 +/- 1.6% and 80 +/- 3.3% of nitrergic neurons in proximal and distal colon respectively. CRF and stressin(1)-A elicit defecation and diarrhoea. These data support that one mechanism through which peripherally injected CRF ligands stimulate colonic function involves a direct action on colonic cholinergic and nitrergic myenteric neurons expressing CRF(1) receptor.


Assuntos
Fibras Colinérgicas/metabolismo , Colo/inervação , Hormônio Liberador da Corticotropina/análogos & derivados , Hormônio Liberador da Corticotropina/farmacologia , Plexo Mientérico/metabolismo , Neurônios Nitrérgicos/metabolismo , Peptídeos Cíclicos/farmacologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Animais , Fibras Colinérgicas/patologia , Colo/patologia , Motilidade Gastrointestinal/efeitos dos fármacos , Masculino , Plexo Mientérico/patologia , Neurônios Nitrérgicos/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Hormônio Liberador da Corticotropina/agonistas
8.
Peptides ; 27(6): 1376-82, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16337713

RESUMO

The effects of intravenous (iv) adrenomedullin (AM) on gastric emptying were investigated in conscious rats. AM induced a maximal 50% inhibition of gastric emptying at a dose of 1.2 nmol/kg. AM was about two-fold less potent than alpha-calcitonin gene-related peptide (alpha-CGRP), which induced a similar 50% maximal inhibition of gastric emptying at 0.6 nmol/kg. Delayed gastric emptying induced by i.v. AM and alpha-CGRP was prevented by peripheral injection of the selective CGRP1 antagonist, CGRP8-37, and by pretreatment with indomethacin, while not altered by blockade of the sympathetic nervous system with propranolol. These data indicate that peripheral AM inhibits gastric emptying through the interaction with CGRP8-37 -sensitive receptors, likely CGRP1 receptors, and the recruitment of prostaglandin-dependent mechanisms.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Esvaziamento Gástrico/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Prostaglandinas/metabolismo , Adrenomedulina , Animais , Peptídeo Relacionado com Gene de Calcitonina/química , Humanos , Indometacina/farmacologia , Masculino , Fragmentos de Peptídeos/química , Peptídeos/química , Peptídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos beta/metabolismo , Receptores da Calcitonina/metabolismo , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/metabolismo
9.
Peptides ; 27(11): 2811-9, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16934368

RESUMO

Obestatin is a new peptide for which anorexigenic effects were recently reported in mice. We investigate whether peripheral injection of obestatin or co-injection with cholecystokinin (CCK) can modulate food intake, gastric motor function (intragastric pressure and emptying) and gastric vagal afferent activity in rodents. Obestatin (30, 100 and 300 microg/kg, i.p.) did not influence cumulative food intake for the 2h post-injection in rats or mice nor gastric emptying in rats. In rats, obestatin (300 microg/kg) did not modify CCK (1 microg/kg, i.p.)-induced significant decrease in food intake (36.6%) and gastric emptying (31.0%). Furthermore, while rats injected with CCK (0.3 microg/kg, i.v.) displayed gastric relaxation, no change in gastric intraluminal pressure was elicited by obestatin (300 microg/kg, i.v.) pre- or post-CCK administration. In in vitro rat gastric vagal afferent preparations, 20 units that had non-significant changes in basal activity after obestatin at 30 microg responded to CCK at 10 ng by a 182% increase. These data show that obestatin neither influences cumulative food intake, gastric motility or vagal afferent activity nor CCK-induced satiety signaling.


Assuntos
Colecistocinina/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Esvaziamento Gástrico/efeitos dos fármacos , Hormônios Peptídicos/farmacologia , Resposta de Saciedade/efeitos dos fármacos , Nervo Vago/efeitos dos fármacos , Animais , Interações Medicamentosas , Esvaziamento Gástrico/fisiologia , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Estômago/efeitos dos fármacos , Estômago/inervação , Estômago/fisiologia , Nervo Vago/fisiologia
10.
Auton Neurosci ; 125(1-2): 42-52, 2006 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-16520096

RESUMO

Pavlov's pioneering work established that sham-feeding induced by sight or smell of food or feeding in dogs with permanent esophagostomy stimulates gastric acid secretion through vagal pathways. Brain circuitries and transmitters involved in the central vagal regulation of gastric function have recently been unraveled. Neurons in the dorsal vagal complex including the dorsal motor nucleus of the vagus (DMN) express thyrotropin-releasing hormone (TRH) receptor and are innervated by TRH fibers originating from TRH synthesizing neurons in the raphe pallidus, raphe obscurus and the parapyramidal regions. TRH injected into the DMN or cisterna magna increases the firing of DMN neurons and gastric vagal efferent discharge, activates cholinergic neurons in gastric submucosal and myenteric plexuses, and induces a vagal-dependent, atropine-sensitive stimulation of gastric secretory (acid, pepsin) and motor functions. TRH antibody or TRH-R1 receptor oligodeoxynucleotide antisense pretreatment in the cisterna magna or DMN abolished vagal-dependent gastric secretory and motor responses to sham-feeding, 2-deoxy-D-glucose, cold exposure and chemical activation of cell bodies in medullary raphe nuclei. TRH excitatory action in the DMN is potentiated by co-released prepro-TRH-(160-169) flanking peptide, Ps4 and 5-HT, and inhibited by a number of peptides involved in the stress/immune response and inhibition of food-intake. These neuroanatomical, electrophysiological and neuropharmacological data are consistent with a physiological role of brainstem TRH in the central vagal stimulation of gastric myenteric cholinergic neurons in response to several vagal dependent stimuli including sham-feeding.


Assuntos
Tronco Encefálico/metabolismo , Comportamento Alimentar/fisiologia , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Estômago/inervação , Nervo Vago/metabolismo , Acetilcolina/metabolismo , Animais , Tronco Encefálico/anatomia & histologia , Humanos , Vias Neurais/anatomia & histologia , Vias Neurais/metabolismo , Placebos , Estômago/fisiologia
11.
Neurogastroenterol Motil ; 27(1): 1-6, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25557223

RESUMO

Corticotropin-releasing factor (CRF)-CRF1 receptor in the brain plays a key role in stress-related alterations of behavior including anxiety/depression, and autonomic and visceral functions. In particular, CRF1 signaling mediates hypersensitivity to colorectal distension (CRD) in various models (early life adverse events, repeated psychological stress, chronic high anxiety, postcolonic inflammation, or repeated nociceptive CRD). So far, knowledge of brain sites involved is limited. A recent article demonstrates in rats that CRF microinjected into the central amygdala (CeA) induces a hyperalgesic response to CRD and enhances the noradrenaline and dopamine levels at this site. The visceral and noradrenaline, unlike dopamine, responses were blocked by a CRF1 antagonist injected into the CeA. Here, we review the emerging role that CRF-CRF1 signaling plays in the CeA to induce visceral hypersensitivity. In the somatic pain field, CRF in the CeA was shown to induce pain sensitization. This is mediated by the activation of postsynaptic CRF1 receptors and protein kinase A signaling that increases N-methyl-d-aspartate receptor neurotransmission. In addition, the activation of tetraethylamonium-sensitive ion channels such as Kv3 accelerates repolarization and firing rate. Whether facilitation of pain transmission underlies CRF action in the CeA-induced visceral hypersensitivity will need to be delineated. CRF1 signaling in the CeA is also an important component of the neuronal circuitry inducing anxiety-like behavior and positioned at the interphase of the reciprocal relationship between pain and affective state. The hyperactivity of this system may represent the neuroanatomical and biochemical substrate contributing to the coexpression of hypersensitivity to CRD and mood disorders in subsets of irritable bowel syndrome patients.


Assuntos
Núcleo Central da Amígdala/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Hiperalgesia/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Animais , Ansiedade/metabolismo , Colo/metabolismo , Dilatação Patológica , Humanos , Camundongos , Neurônios/metabolismo , Dor , Ratos , Reto/metabolismo , Transdução de Sinais , Estresse Psicológico/metabolismo
12.
Endocrinology ; 105(3): 660-5, 1979 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-467325

RESUMO

Bombesin acts within the brain to produce a prompt and sustained hyperglycemia, hyperglucagonemia, and relative or absolute hypoinsulinemia. Bombesin does not decrease plasma glucose turnover. Acute adrenalectomy but not hypophysectomy prevents hyperglycemia and hyperglucagonemia after intracisternal administration of bombesin. Administration of bombesin into the lateral ventricle of awake, unrestrained animals results in elevation of plasma glucose, preceded by a significant increase in plasma epinephrine and no increase in plasma norepinephrine or dopamine. Systemic administration of somatostatin prevents bombesin-induced hyperglycemia and hyperglucagonemia. These data support the conclusion that bombesin acts within the brain to increase sympathetic outflow resulting in increased adrenalmedullary epinephrine secretion, followed by depression of plasma insulin and elevation of plasma glucagon and glucose.


Assuntos
Bombesina/farmacologia , Encéfalo/efeitos dos fármacos , Epinefrina/metabolismo , Hiperglicemia/induzido quimicamente , Peptídeos/farmacologia , Glândulas Suprarrenais/fisiologia , Animais , Glicemia/metabolismo , Glucagon/sangue , Insulina/sangue , Fígado/metabolismo , Masculino , Hipófise/fisiologia , Ratos , Somatostatina/farmacologia
13.
Endocrinology ; 105(1): 220-4, 1979 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-109288

RESUMO

The effects of bombesin and other unrelated oligopeptides on hormonal changes induced by stress were studied in conscious adult male rats. Restraint in the cold for 1 h increased plasma corticosterone and PRL levels and decreased GH values but had no effect on LH levels. Bombesin (5 microgram), given intracerebroventricularly (ivt) before stress, inhibited the PRL rise without affecting corticosterone, GH, or LH response. A complete blockade of PRL rise was observed with doses of bombesin ranging from 5 microgram to 100 ng ivt, regardless of the duration (15, 30, 45, or 60 min) or the nature (cold exposure or restraint at room temperature) of the stressor agents. Bombesin was 10(3) more potent as a PRL inhibitor when given ivt than when given iv, and its ivt effect was not reversed by naloxone (1 or 10 mg/kg). Among other unrelated peptides tested (beta-endorphin, neurotensin, substance P, and TRH; 5 microgram ivt), only neurotensin decreased plasma PRL levels in rats subjected to restraint in the cold for 1 h. These results show that in conscious male rats, centrally administered bombesin has a very potent and long acting inhibitory effect on PRL release induced by acute stress. Since a bombesin-like peptide has been found in rat brain, its physiological role in PRL regulation remains to be elucidated.


Assuntos
Bombesina/farmacologia , Neurotensina/farmacologia , Peptídeos/farmacologia , Hormônios Adeno-Hipofisários/metabolismo , Prolactina/metabolismo , Estresse Fisiológico/fisiopatologia , Animais , Temperatura Baixa , Corticosterona/metabolismo , Endorfinas/farmacologia , Hormônio do Crescimento/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Ratos , Somatostatina/farmacologia , Substância P/farmacologia , Hormônio Liberador de Tireotropina/farmacologia
14.
Endocrinology ; 138(9): 3749-55, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9275061

RESUMO

The central action of human adrenomedullin (AM) to influence gastric emptying and the peripheral mechanisms involved were studied in conscious rats. The 20-min rate of gastric emptying of a methylcellulose solution was assessed after intracisternal (i.c.) injection of AM or rat alpha-calcitonin gene-related peptide (alphaCGRP). AM and alphaCGRP dose-dependently inhibited gastric emptying with i.c. ED50 values of 120 and 100 pmol, respectively. Human proadrenomedullin N-terminal 20 peptide (150-600 pmol, i.c.) and AM (150 pmol, i.v.) had no effect. The inhibitory actions of AM and alphaCGRP (150 pmol, i.c.) were completely blocked by the CGRP antagonist, human CGRP-(8-37) injected i.c. at 30 microg, but not at 15 microg. The CRF antagonist, [D-Phe12,Nle(21,38),C(alpha)MeLeu37]CRF-(12-41) (10 microg/rat) injected i.c. prevented i.c. rat/human CRF (150 pmol)-induced 53% inhibition of gastric emptying while not modifying the effect of AM. The action of AM (150 pmol, i.c.) was abolished by bilateral adrenalectomy or the beta-adrenergic blocker, propranolol (1 mg/kg, i.p.), but was not altered by indomethacin (5 mg/kg, i.p.) or subdiaphragmatic vagotomy. These results indicate that i.c. AM and alphaCGRP equipotently inhibit gastric emptying through mechanisms similarly antagonized by a high dose of CGRP-(8-37). The central AM action is mediated through adrenal-dependent, beta-adrenergic pathways independently from activation of central CRF receptors.


Assuntos
Esvaziamento Gástrico/efeitos dos fármacos , Peptídeos/farmacologia , Adrenalectomia , Antagonistas Adrenérgicos beta/farmacologia , Adrenomedulina , Animais , Peptídeo Relacionado com Gene de Calcitonina/antagonistas & inibidores , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Hormônio Liberador da Corticotropina/antagonistas & inibidores , Hormônio Liberador da Corticotropina/farmacologia , Humanos , Indometacina/farmacologia , Cinética , Masculino , Metilcelulose/metabolismo , Fragmentos de Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Vagotomia
15.
Endocrinology ; 97(3): 731-4, 1975 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1175518

RESUMO

PCN (3beta-Hydroxy-20-oxo-pregn-5-ene-16alpha-carbonitrile), a potent catatoxic steroid devoid of all other classic hormonal properties, given in doses amply sufficient to induce hepatic drug-metabolizing enzymes and inhibit the anesthesia caused by massive amounts of progesterone in pregnant rats, did not modify: 1. the pregnant:mated ratio, 2. the length of gestation, 3. the number of implantation sites, and 4. the size of the litter. The viability and postpartum development of the offspring were normal, as judged by gross macroscopic examination. PCN had no adverse effects on pregnancy after hypophysectomy (day 12), or ovariectomy (day 8) in rats treated with maintenance doses of progesterone and estrone. It would therefore appear that the pregnancy-maintaining capacity of endogenous or exogenous steroids (administered as substitution therapy following the glandular extirpations) was not altered by PCN and that hypothalamo-hypophyseal regulatory mechanisms were not responsible for the failure of the steroid to provoke abortion. As judged by our in vivo experiment, this pure hepatic microsomal exzyme inducer does not modify the steady state of steroid-dependent physiologic functions.


Assuntos
Abortivos , Prenhez/efeitos dos fármacos , Carbonitrila de Pregnenolona/farmacologia , Animais , Castração , Estrona/farmacologia , Feminino , Hipofisectomia , Gravidez , Progesterona/farmacologia , Ratos
16.
Endocrinology ; 140(1): 43-9, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9886805

RESUMO

Medullary TRH regulates autonomic activity, and altered thyroid status is associated with autonomic disorders. We investigated whether thyroid hormone exerts a negative feedback regulation on TRH gene expression in the medullary caudal raphe nuclei. Medullary pro-TRH messenger RNAs (mRNAs) were mainly located in the raphe pallidus and raphe obscurus neurons as shown by in situ hybridization and were significantly increased by 70% and 160-230% by Northern blot analyses in 24 h fasted rats at 1 and 3-5 weeks after thyroidectomy, respectively, when serum T4 levels were reduced by 75-87%. The increased pro-TRH mRNA on the 30th day after thyroidectomy was reversed to euthyroid levels by T4 replacement (2 or 4 microg/100 g x day). T4 injections (10 or 100 microg/100 g x day for 30 days) did not significantly influence medullary pro-TRH mRNA levels in sham-operated rats. Thyroidectomized rats fed normally showed a 500% increase in pro-TRH mRNA levels 30 days after the surgery, while those fasted for 24 h showed only a 180% increase. These data indicate that medullary TRH gene expression is enhanced during hypothyroidism due to the lack of negative feedback regulation by thyroid hormone, and this response is modulated by feeding state. These findings may have important relevance to understanding autonomic-related visceral alterations induced by hypothyroidism.


Assuntos
Regulação da Expressão Gênica/fisiologia , Núcleos da Rafe/fisiologia , Hormônio Liberador de Tireotropina/genética , Animais , Peso Corporal , Retroalimentação/fisiologia , Hipotireoidismo/genética , Hibridização In Situ , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Glândula Tireoide/fisiologia , Tireoidectomia , Hormônio Liberador de Tireotropina/biossíntese , Tiroxina/sangue
17.
Endocrinology ; 139(9): 3730-5, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9724024

RESUMO

Cold exposure increases TRH gene expression in hypothalamic and raphe nuclei and results in a vagal activation of gastric function. We investigated the role of medullary TRH receptors in cold (4-6 C, 90 min)-induced stimulation of gastric motor function in fasted conscious rats using intracisternal injections of TRH receptor (TRHr) antisense oligodeoxynucleotides (100 microg twice, -48 and -24 h). The gastric emptying of a methyl-cellulose solution was assessed by the phenol red method. TRH (0.1 microg) or the somatostatin subtype 5-preferring analog, BIM-23052 (1 microg), injected intracisternally increased basal gastric emptying by 34% and 47%, respectively. TRHr antisense, which had no effect on basal emptying, blocked TRH action but did not influence that of BIM-23052. Cold exposure increased gastric emptying by 64%, and the response was inhibited by vagotomy, atropine (0.1 mg/kg, i.p.), and TRHr antisense (intracisternally). Saline or mismatched oligodeoxynucleotides, injected intracisternally under similar conditions, did not alter the enhanced gastric emptying induced by cold or intracisternal injection of TRH or BIM-23052. These results indicate that TRH receptor activation in the brain stem mediates acute cold-induced vagal cholinergic stimulation of gastric transit, and that medullary TRH may play a role in the autonomic visceral responses to acute cold.


Assuntos
Cisterna Magna/fisiologia , Temperatura Baixa , Esvaziamento Gástrico/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Receptores do Hormônio Liberador da Tireotropina/genética , Nervo Vago/fisiologia , Animais , Atropina/farmacologia , Masculino , Oligonucleotídeos Antissenso/genética , Ratos , Ratos Sprague-Dawley , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Hormônio Liberador de Tireotropina/farmacologia , Fatores de Tempo , Vagotomia
18.
Endocrinology ; 100(5): 1391-3, 1977 May.
Artigo em Inglês | MEDLINE | ID: mdl-403068

RESUMO

The concentration of thyrotropin-releasing hormone (TRH) was measured by specific radioimmunoassay in three different brain regions of rats sacrificed every 4 h over a 24 h period. TRH concentration reached a zenith at 1200 h both in the hypothalamus and the amygdala but did not vary in the forebrain. These results indicate that a nyctohemaral variation of TRH levels exists in hypothalamic and extrahypothalamic brain areas.


Assuntos
Encéfalo/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Tonsila do Cerebelo/metabolismo , Animais , Ritmo Circadiano , Diencéfalo/metabolismo , Hipotálamo/metabolismo , Masculino , Ratos , Telencéfalo/metabolismo
19.
Endocrinology ; 109(4): 1254-8, 1981 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-6269839

RESUMO

In rats, chronic intermittent immobilization stress induced a drastic fall in the plasma concentration and testicular content of testosterone (T) without detectable changes in plasma LH values. In vitro basal T production by interstitial cell-enriched preparations from stressed rats and the responses to hCG, dibutyryl cAMP, or choleratoxin were suppressed, while cAMP production was not modified. The increase in plasma T concentrations in control animals was identical after the in vivo injection of 5, 10, or 50 IU hCG, while stressed rats failed to respond to 5 IU, but showed a response similar to that of control animals with 10 and 50 IU. These results suggest that chronic intermittent immobilization stress decreases Leydig cell sensitivity to gonadotropins.


Assuntos
Estresse Fisiológico/metabolismo , Testículo/metabolismo , Testosterona/metabolismo , Animais , Bucladesina/farmacologia , Toxina da Cólera/farmacologia , Gonadotropina Coriônica/farmacologia , AMP Cíclico/metabolismo , Cinética , Hormônio Luteinizante/sangue , Masculino , Ratos , Ratos Endogâmicos , Restrição Física , Testículo/efeitos dos fármacos
20.
J Comp Neurol ; 349(2): 212-22, 1994 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-7860779

RESUMO

Previous neuropharmacological studies indicate that brain peptides are involved in mediating gastric stasis induced by abdominal surgery. Central pathways activated by abdominal surgery were investigated in the rat by using Fos protein as a marker of neuronal activation. Abdominal surgery (laparotomy alone or combined with cecal manipulation) was performed under brief enflurane anesthesia (7-8 minutes), and 1 hour later rats were killed and brains processed for Fos immunoreactivity. Double labeling with Fos and arginine vasopressin, oxytocin, or tyrosine hydroxylase antibodies was also performed. Abdominal surgery induced Fos staining in the nucleus tractus solitarii, paraventricular and supraoptic nuclei of the hypothalamus, locus coeruleus, and ventrolateral medulla. After abdominal surgery, 18-25% of vasopressin and 18-33% of oxytocin-labeled cells were found to be Fos positive in the paraventricular nucleus and 15% of activated cells in the nucleus tractus solitarii were positive for tyrosine hydroxylase immunoreactivity. Enflurane alone induced c-fos expression in the same brain area; however, the number of Fos-positive cells and double-labeled cells were decreased two- to fivefold and three- to eightfold, respectively, compared with the abdominal surgery groups. These data show that abdominal surgery induced activation of specific hypothalamic, pontine, and medullary neurons. These findings may have implications for the understanding of central mechanisms involved in mediating gastric ileus following abdominal surgery.


Assuntos
Abdome/cirurgia , Química Encefálica/fisiologia , Proteínas Proto-Oncogênicas c-fos/análise , Animais , Biomarcadores/química , Mapeamento Encefálico , Técnicas Imunoenzimáticas , Masculino , Proteínas do Tecido Nervoso/análise , Vias Neurais/química , Núcleo Hipotalâmico Paraventricular/química , Ratos , Ratos Sprague-Dawley , Núcleo Solitário/química , Núcleo Supraóptico/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA