Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 216
Filter
1.
Br J Pharmacol ; 164(8): 1959-75, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21627635

ABSTRACT

BACKGROUND AND PURPOSE: Infusion of corticotropin-releasing factor (CRF)/urocortin (Ucn) family peptides suppresses feeding in mice. We examined whether rats show peripheral CRF/Ucn-induced anorexia and determined its behavioural and pharmacological bases. EXPERIMENTAL APPROACH: Male Wistar rats (n= 5-12 per group) were administered (i.p.) CRF receptor agonists with different subtype affinities. Food intake, formation of conditioned taste aversion and corticosterone levels were assessed. In addition, Ucn 1- and Ucn 2-induced anorexia was studied in fasted CRF(2) knockout (n= 11) and wild-type (n= 13) mice. KEY RESULTS: Ucn 1, non-selective CRF receptor agonist, reduced food intake most potently (~0.32 nmol·kg(-1) ) and efficaciously (up to 70% reduction) in fasted and fed rats. The peptides' rank-order of anorexic potency was Ucn 1 ≥ Ucn 2 > >stressin(1) -A > Ucn 3, and efficacy, Ucn 1 > stressin(1) -A > Ucn 2 = Ucn 3. Ucn 1 reduced meal frequency and size, facilitated feeding bout termination and slowed eating rate. Stressin(1) -A (CRF(1) agonist) reduced meal size; Ucn 2 (CRF(2) agonist) reduced meal frequency. Stressin(1) -A and Ucn 1, but not Ucn 2, produced a conditioned taste aversion, reduced feeding efficiency and weight regain and elicited diarrhoea. Ucn 1, but not Ucn 2, also increased corticosterone levels. Ucn 1 and Ucn 2 reduced feeding in wild-type, but not CRF(2) knockout, mice. CONCLUSIONS AND IMPLICATIONS: CRF(1) agonists, Ucn 1 and stressin(1) -A, reduced feeding and induced interoceptive stress, whereas Ucn 2 potently suppressed feeding via a CRF(2) -dependent mechanism without eliciting malaise. Consistent with their pharmacological differences, peripheral urocortins have diverse effects on appetite.


Subject(s)
Corticotropin-Releasing Hormone/analogs & derivatives , Feeding Behavior , Peptides, Cyclic/physiology , Receptors, Corticotropin-Releasing Hormone/physiology , Stress, Physiological , Urocortins/physiology , Animals , Corticosterone/blood , Corticotropin-Releasing Hormone/physiology , Female , Male , Mice , Mice, Knockout , Rats , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/genetics
2.
Diabetologia ; 54(9): 2392-403, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21667214

ABSTRACT

AIMS/HYPOTHESIS: Urocortins are the endogenous ligands for the corticotropin-releasing factor receptor type 2 (CRFR2), which is implicated in regulating energy balance and/or glucose metabolism. We determined the effects of chronic CRFR2 activation on metabolism in vivo, by generating and phenotyping transgenic mice overproducing the specific CRFR2 ligand urocortin 3. METHODS: Body composition, glucose metabolism, insulin sensitivity, energy efficiency and expression of key metabolic genes were assessed in adult male urocortin 3 transgenic mice (Ucn3(+)) under control conditions and following an obesogenic high-fat diet (HFD) challenge. RESULTS: Ucn3(+) mice had increased skeletal muscle mass with myocyte hypertrophy. Accelerated peripheral glucose disposal, increased respiratory exchange ratio and hypoglycaemia on fasting demonstrated increased carbohydrate metabolism. Insulin tolerance and indices of insulin-stimulated signalling were unchanged, indicating these effects were not mediated by increased insulin sensitivity. Expression of the transgene in Crfr2 (also known as Crhr2)-null mice negated key aspects of the Ucn3(+) phenotype. Ucn3(+) mice were protected from the HFD-induced hyperglycaemia and increased adiposity seen in control mice despite consuming more energy. Expression of uncoupling proteins 2 and 3 was higher in Ucn3(+) muscle, suggesting increased catabolic processes. IGF-1 abundance was upregulated in Ucn3(+) muscle, providing a potential paracrine mechanism in which urocortin 3 acts upon CRFR2 to link the altered metabolism and muscular hypertrophy observed. CONCLUSIONS/INTERPRETATION: Urocortin 3 acting on CRFR2 in skeletal muscle of Ucn3(+) mice results in a novel metabolically favourable phenotype, with lean body composition and protection against diet-induced obesity and hyperglycaemia. Urocortins and CRFR2 may be of interest as potential therapeutic targets for obesity.


Subject(s)
Dietary Fats/adverse effects , Hyperglycemia/metabolism , Hyperglycemia/prevention & control , Obesity/metabolism , Obesity/prevention & control , Urocortins/genetics , Urocortins/metabolism , Animals , Body Composition/drug effects , Body Composition/physiology , Dietary Fats/pharmacology , Disease Models, Animal , Energy Metabolism/drug effects , Energy Metabolism/physiology , Glucose/metabolism , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Phenotype , Receptors, Corticotropin-Releasing Hormone/deficiency , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism
3.
Endocrinology ; 152(1): 138-50, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21106875

ABSTRACT

Urocortin 3 (Ucn 3), member of the corticotropin-releasing factor (CRF) family of peptide hormones, is released from ß-cells to potentiate insulin secretion. Ucn 3 activates the CRF type-2 receptor (CRFR2) but does not activate the type-1 receptor (CRFR1), which was recently demonstrated on ß-cells. While the direct actions of Ucn 3 on insulin secretion suggest the presence of cognate receptors within the islet microenvironment, this has not been established. Here we demonstrate that CRFR2α is expressed by MIN6 insulinoma cells and by primary mouse and human islets, with no detectable expression of CRFR2ß. Furthermore, stimulation of MIN6 cells or primary mouse islets in vitro or in vivo with glucocorticoids (GCs) robustly and dose-dependently increases the expression of CRFR2α, while simultaneously inhibiting the expression of CRFR1 and incretin receptors. Luciferase reporters driven by the mouse CRFR1 or CRFR2α promoter in MIN6 cells confirm these differential effects of GCs. In contrast, GCs inhibit CRFR2α promoter activity in HEK293 cells and inhibit the expression of CRFR2ß in A7r5 rat aortic smooth muscle cells and differentiated C2C12 myotubes. These findings suggest that the GC-mediated increase of CRFR2α depends on the cellular context of the islet and deviates from the GC-mediated suppression of CRFR1 and incretin receptors. Furthermore, GC-induced increases in CRFR2α expression coincide with increased Ucn 3-dependent activation of cAMP and MAPK pathways. We postulate that differential effect of GCs on the expression of CRFR1 and CRFR2α in the endocrine pancreas represent a mechanism to shift sensitivity from CRFR1 to CRFR2 ligands.


Subject(s)
Corticosterone/pharmacology , Glucocorticoids/pharmacology , Insulinoma/metabolism , Islets of Langerhans/drug effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Cell Line, Tumor , Drug Implants , Humans , Insulinoma/drug therapy , Islets of Langerhans/metabolism , Mice , Mifepristone , Protein Isoforms , Rats , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/genetics
4.
Oncogene ; 28(24): 2324-36, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19421146

ABSTRACT

Cripto is a developmental oncoprotein that signals via mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K)/Akt and Smad2/3 pathways. However, the molecular basis for Cripto coupling to these pathways during embryogenesis and tumorigenesis is not fully understood. In this regard, we recently demonstrated that Cripto forms a cell surface complex with the HSP70 family member glucose-regulated protein-78 (GRP78). Here, we provide novel functional evidence demonstrating that cell surface GRP78 is a necessary mediator of Cripto signaling in human tumor, mammary epithelial and embryonic stem cells. We show that targeted disruption of the cell surface Cripto/GRP78 complex using shRNAs or GRP78 immunoneutralization precludes Cripto activation of MAPK/PI3K pathways and modulation of activin-A, activin-B, Nodal and transforming growth factor-beta1 signaling. We further demonstrate that blockade of Cripto binding to cell surface GRP78 prevents Cripto from increasing cellular proliferation, downregulating E-Cadherin, decreasing cell adhesion and promoting pro-proliferative responses to activin-A and Nodal. Thus, disrupting the Cripto/GRP78 binding interface blocks oncogenic Cripto signaling and may have important therapeutic value in the treatment of cancer.


Subject(s)
Epidermal Growth Factor/metabolism , Heat-Shock Proteins/metabolism , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Smad Proteins/metabolism , Activins/pharmacology , Antibodies/pharmacology , Blotting, Western , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum Chaperone BiP , Epidermal Growth Factor/genetics , GPI-Linked Proteins , Heat-Shock Proteins/genetics , Heat-Shock Proteins/immunology , Humans , Immunoprecipitation , Intercellular Signaling Peptides and Proteins , Luciferases/genetics , Luciferases/metabolism , Membrane Glycoproteins/genetics , Microscopy, Confocal , Models, Biological , Neoplasm Proteins/genetics , Protein Binding/drug effects , RNA, Small Interfering/genetics , Radioligand Assay , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism
5.
Neuroscience ; 162(1): 5-13, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19358876

ABSTRACT

Defeat is a social stressor involving subordination by a threatening conspecific. Type 2 corticotropin-releasing factor receptors (CRF(2)) are abundant in brain regions implicated in defeat responses and are putative stress-related molecules. The present study sought to determine whether neuroactivation and CRF(2) expression co-occurred at brain region or cellular levels following acute defeat. Male "intruder" Wistar rats were placed into the cage of an aggressive "resident" Long-Evans rat (n=6). Upon defeat, intruders (n=6) were placed in a wire-mesh chamber and were returned to the resident's cage for an additional 75 min. Controls (n=6) were handled and returned to their home cage for the same duration. Coronal brain sections were stained for an immediate early gene product, Fos, as a neuronal activation marker. Combined immunohistochemistry with in situ hybridization was performed on a subset of brain sections from defeated intruders to visualize Fos immunoreactivity and CRF(2) mRNA jointly. Defeated rats had fivefold, sevenfold, and 10-fold more Fos-positive cells than controls in the arcuate, ventromedial nucleus of the hypothalamus, and medial amygdala post-defeat. Significant colocalization of CRF(2) mRNA and Fos-positive cells was observed in the posterior medial amygdala but not in the arcuate nucleus or ventromedial hypothalamus. The results indicate CRF(2) receptor-positive neurons in the posterior medial amygdala are involved in the neural response to social defeat.


Subject(s)
Amygdala/metabolism , Dominance-Subordination , Neurons/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Stress, Psychological/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Prosencephalon/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Rats , Rats, Long-Evans , Rats, Wistar , Ventromedial Hypothalamic Nucleus/metabolism
6.
Eur J Neurosci ; 26(8): 2303-14, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17953621

ABSTRACT

The actions of corticotropin-releasing factor (CRF) and related peptides are mediated by two receptors (CRF(1) and CRF(2)). The respective role of each subtype in the control of food intake remains poorly known. In the present study, we examined the quantity and microstructure of ingestive behavior of knockout (KO) mice lacking CRF(2) receptors and their wild-type (WT) littermates. Under basal conditions, CRF(2) KO mice showed increased nocturnal food intake, evident as an increased zenith in circadian cosinor analysis of food intake. Microstructure analysis revealed that this greater food intake reflected increased meal size, rather than meal frequency, suggesting a decreased satiating value of food. Following acute restraint stress, CRF(2) KO mice showed an intact immediate anorectic response with increased latency to eat and decreased meal size. However, CRF(2) deletion abolished the prolonged phase of restraint-induced anorexia. CRF(2) KO mice did not differ from WT controls in feeding responses to food deprivation or injection of ghrelin receptor agonists. Independent of genotype, food deprivation increased food intake, with dramatic changes in meal size, meal frequency, water : food ratio and eating rate. Acyl-ghrelin or BIM-28131, a potent ghrelin analog, dose-dependently stimulated food intake by increasing meal size (ghrelin, BIM-28131) and meal number (BIM-28131), while slowing the average eating rate (BIM-28131) similarly in WT and KO mice. These results suggest that the CRF(2) receptor is involved in the control of meal size during the active phase of eating and following acute exposure to stress.


Subject(s)
Eating/physiology , Feeding Behavior/psychology , Receptors, Corticotropin-Releasing Hormone/physiology , Animals , Behavior, Animal , Dose-Response Relationship, Drug , Drinking/physiology , Eating/drug effects , Eating/genetics , Feeding Behavior/drug effects , Food Deprivation/physiology , Ghrelin/agonists , Ghrelin/analogs & derivatives , Ghrelin/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Periodicity , Receptors, Corticotropin-Releasing Hormone/deficiency , Restraint, Physical/methods , Time Factors
7.
J Pharmacol Exp Ther ; 323(3): 846-54, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17855476

ABSTRACT

The corticotropin-releasing factor (CRF) system mediates stress responses. Extrahypothalamic CRF1 receptor activation has anxiogenic-like properties, but anxiety-related functions of CRF2 receptors remain unclear. The present study determined the effects of intracerebroventricular administration of a CRF2 agonist, urocortin 3, on behavior of male Wistar rats in the shock-probe, social interaction, and defensive withdrawal tests of anxiety-like behavior. Equimolar doses of stressin1-A, a novel CRF1 agonist, were administered to separate rats. The effects of pyrazolo[1,5-a]-1,3,5-triazin-4-amine,8-[4-(bromo)-2-chlorophenyl]-N, N-bis(2-methoxyethyl)-2,7-dimethyl-(9Cl) (MJL-1-109-2), a CRF1 antagonist, on behavior in the shock-probe test also were studied. Stressin1-A increased anxiety-like behavior in the social interaction and shock-probe tests. Stressin1-A elicited behavioral activation and defensive burying at lower doses (0.04 nmol), but it increased freezing, grooming, and mounting at 25-fold higher (1-nmol) doses. Conversely, systemic administration of MJL-1-109-2 (10 mg/kg) had anxiolytic-like effects in the shock-probe test. Unlike stressin1-A or MJL-1-109-2, i.c.v. urocortin 3 infusion did not alter anxiety-like behavior in the shock-probe test across a range of doses that reduced locomotion and rearing and increased grooming. Urocortin 3 also did not decrease social interaction, but it decreased anxiety-like behavior in the defensive withdrawal test at a 2-nmol dose. Thus, i.c.v. administration of CRF1 and CRF2 agonists produced differential, but not opposite, effects on anxiety-like behavior. Urocortin 3 (i.c.v.) did not consistently decrease or increase anxiety-like behavior, the latter unlike effects seen previously after local microinjection of CRF2 agonists into the septum or raphe. With increasing CRF1 activation, however, the behavioral expression of anxiety qualitatively changes from "coping" to "noncoping" and offensive, agonistic behaviors.


Subject(s)
Anxiety/drug therapy , Behavior, Animal/drug effects , Receptors, Corticotropin-Releasing Hormone/agonists , Animals , Anxiety/metabolism , Anxiety/physiopathology , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/analogs & derivatives , Corticotropin-Releasing Hormone/pharmacology , Corticotropin-Releasing Hormone/therapeutic use , Injections, Intraventricular , Male , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Rats , Rats, Wistar , Social Behavior , Triazines/administration & dosage , Triazines/pharmacology , Triazines/therapeutic use , Urocortins/administration & dosage , Urocortins/pharmacology , Urocortins/therapeutic use
8.
Neuroscience ; 128(1): 111-9, 2004.
Article in English | MEDLINE | ID: mdl-15450358

ABSTRACT

We had previously demonstrated that continual-hypoxia stimulated corticotropin-releasing factor (CRF)mRNA in hypothalamus, and release of CRF, as well as enhancing plasma adrenocorticotropic-hormone and corticosterone of rats. The present study demonstrates using in situ autoradiography that CRF receptor 1 (CRFR1) and CRF receptor 2 (CRFR2) mRNA in the rat anterior pituitary is changed by intermittent hypoxia, cold, restraint, alone and in combination. Rats were exposed to intermittent hypoxia for 4 h/day during various periods in a hypobaric chamber. Hypoxia equivalent to an altitude of around 2 km (16.0% O2) or 5 km (10.8% O2) caused a biphasic change in both CRFR1 and R2 mRNA, there being an initial significant decline on day 1 and then an enhancement by day 2. The increase of both receptor subtypes mRNA was relatively well maintained up to 15 days in rats exposed to 2 km intermittently. CRFR2 mRNA in rats exposed to 5 km, after peaking at day 2 therefore declined and was not different to controls at 15 days. Five kilometer hypoxia markedly reduced body weight gain. The increased CRFR1 mRNA was also induced by restraint alone, hypoxia+restraint and hypoxia+cold but not by cold alone. The CRFR2 mRNA was significantly increased by all the stresses except for hypoxia+restraint. These results show that the acute response to intermittent hypoxia is a decrease in the CRF receptor mRNA whereas longer exposure to the three environmental stressors hypoxia, cold and restraint is needed to provoke an increase. This may have important consequences for adaptation to high altitude. The significant differences between the expression of CRFR1 mRNA and CRFR2 mRNA in response to the different stimuli might suggest that the two receptors in the pituitary play different roles in behavior.


Subject(s)
Hypoxia/physiopathology , Pituitary Gland, Anterior/metabolism , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Stress, Psychological/physiopathology , Animals , Autoradiography , Cold Temperature , Corticosterone/blood , In Situ Hybridization , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Restraint, Physical , Time Factors
9.
Eur J Neurosci ; 18(2): 373-82, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12887419

ABSTRACT

The influence of urocortin (UCN) on ingestive behaviours and brain neural activity, as measured immunohistochemically by the presence of Fos protein, was determined in mice. Rat UCN was administered by continuous intracerebroventricular (ICV) or subcutaneous (SC) infusion. ICV infusion of UCN (100 ng/h, 14 days) transiently reduced daily food and water intakes (days 1-4) but body weight was reduced from day 2 into the post-infusion period. Sodium intake was reduced from day 3 to the end of infusion. SC infusion of UCN caused similar but smaller reductions in food and water intakes and body weight, without change in sodium intake. In separate experiments, Fos immunoreactivity was increased in several brain nuclei known to be involved in the control of body fluid and energy homeostasis, e.g. central nucleus of the amygdala, median preoptic nucleus, bed nucleus of the stria terminalis and arcuate nucleus. Increased Fos expression was similar for ICV and SC infusions when measured on days 2-3 or 6-7 of infusion. In conclusion, increases of brain activity by UCN may be associated with stimulation of adrenocorticotrophic hormone release and sympathetic nervous activity, but increases may also indicate suppression of ingestive behaviours by stimulating central inhibitory mechanisms located in areas known to control body fluid and energy homeostasis.


Subject(s)
Brain/metabolism , Corticotropin-Releasing Hormone/pharmacology , Eating/drug effects , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Animals , Brain/drug effects , Corticotropin-Releasing Hormone/administration & dosage , Drinking/drug effects , Immunohistochemistry , Injections, Intraventricular , Male , Mice , Mice, Inbred BALB C , Neuroprotective Agents/administration & dosage , Proto-Oncogene Proteins c-fos/drug effects , Sodium , Urocortins
10.
Mol Psychiatry ; 7(9): 967-74, 2002.
Article in English | MEDLINE | ID: mdl-12399950

ABSTRACT

Corticotropin-releasing hormone (CRH) is both a main regulator of the hypothalamic-pituitary-adrenocortical axis and the autonomic nervous system. CRH receptor type 1 (CRHR1)-deficient mice demonstrate alterations in behavior, impaired stress responses with adrenocortical insufficiency and aberrant neuroendocrine development, but the adrenal medulla has not been analyzed in these animals. Therefore we studied the production of adrenal catecholamines, expression of the enzyme responsible for catecholamine biosynthesis neuropeptides and the ultrastructure of chromaffin cells in CRHR1 null mice. In addition we examined whether treatment of CRHR1 null mice with adrenocorticotropic hormone (ACTH) could restore function of the adrenal medulla. CRHR1 null mice received saline or ACTH, and wild-type or heterozygous mice injected with saline served as controls. Adrenal epinephrine levels in saline-treated CRHR1 null mice were 44% those of controls (P<0.001), and the phenylethanolamine N-methyltransferase (PNMT) mRNA levels in CRHR1 null mice were only 25% of controls (P <0.001). ACTH treatment increased epinephrine and PNMT mRNA level in CRHR1 null mice but failed to restore them to normal levels. Proenkephalin mRNA in both saline- and ACTH-treated CRHR1 null mice were higher than in control animals (215.8% P <0.05, 268.9% P <0.01) whereas expression of neuropeptide Y and chromogranin B did not differ. On the ultrastructural level, chromaffin cells in saline-treated CRHR1 null mice exhibited a marked depletion in epinephrine-storing secretory granules that was not completely normalized by ACTH-treatment. In conclusion, CRHR1 is required for a normal chromaffin cell structure and function and deletion of this gene is associated with a significant impairment of epinephrine biosynthesis.


Subject(s)
Chromaffin Cells/pathology , Chromaffin Cells/physiology , Receptors, Corticotropin-Releasing Hormone/genetics , Stress, Physiological/physiopathology , Adrenal Glands/enzymology , Adrenal Glands/pathology , Adrenocorticotropic Hormone/pharmacology , Animals , Behavior, Animal/physiology , Chromaffin Cells/ultrastructure , Chromogranins/genetics , Corticosterone/blood , Enkephalins/genetics , Epinephrine/biosynthesis , Epinephrine/blood , Mice , Mice, Knockout , Microscopy, Electron , Neuropeptide Y/genetics , Phenylethanolamine N-Methyltransferase/genetics , Phenylethanolamine N-Methyltransferase/metabolism , Phosphoproteins/genetics , Protein Precursors/genetics , RNA, Messenger/analysis , Stress, Physiological/metabolism , Stress, Physiological/pathology
11.
Psychopharmacology (Berl) ; 160(2): 113-21, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11875628

ABSTRACT

RATIONALE: Corticotropin-releasing factor (CRF) and urocortin (Ucn) belong to the CRF-related family, share a high degree of structural homology and bind to CRF receptors. However, compared with CRF, Ucn was shown to display either weaker or similar anxiogenic-like effects in vivo. OBJECTIVE: To compare the anxiogenic-like responses of rats injected intracerebroventricularly (ICV) with different doses of either rat/human CRF (r/hCRF) or rat Ucn (rUcn) at different intervals after injection. METHODS: Rats were tested on three validated paradigms of emotional behavior [i.e. elevated plus-maze (EPM), defensive withdrawal (DW) and conflict test (CT)] 5 and 30 min after treatment. RESULTS: In the EPM test only r/hCRF, but not rUcn, produced anxiogenic-like effects at the dose of 1.0 microg, when the peptides were injected 5 min before testing. At 30 min after injection, both peptides caused a significant reduction of open arms exploration, rUcn being effective at 0.01 microg. In the DW test both peptides were equally potent in decreasing the exploratory behavior and increasing the time spent in the chamber at the dose of 1.0 microg when tested 30 min after injection. In the CT both rUcn (0.25-1.0 microg) and r/hCRF (0.75-1.0 microg) decreased significantly the responding in the punished component. However, rUcn reduced food responding also in the unpunished component possibly due to its powerful anorectic activity. CONCLUSIONS: Comparison of anxiogenic-like activities of r/hCRF and rUcn at doses up to 1.0 microg revealed striking differential effects that depended on the time of testing after ICV peptide injection, and on the paradigm of anxiety used. These results suggest that the onset of r/hCRF and rUcn actions related to behavioral responses to anxiety is likely to depend on brain peptide-specific mechanisms including binding properties to CRF-receptors, differential distribution to specific functional brain sites and the distribution and effectiveness of binding-protein interactions.


Subject(s)
Anxiety/chemically induced , Behavior, Animal/drug effects , Corticotropin-Releasing Hormone/adverse effects , Animals , Anxiety/physiopathology , Corticotropin-Releasing Hormone/administration & dosage , Dose-Response Relationship, Drug , Injections, Intraventricular , Male , Rats , Rats, Wistar , Time Factors , Urocortins
12.
Am J Physiol Regul Integr Comp Physiol ; 282(1): R10-8, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11742818

ABSTRACT

Experimental stress and the administration of the stress hormone ACTH have been reported to stimulate sodium appetite in many nonprimate species. Experiments were conducted to determine whether prolonged intracerebroventricular infusions of the neuropeptides corticotropin-releasing factor (CRF) and urocortin (Ucn), or systemic administration of ACTH, affected ingestive behaviors in a nonhuman primate, the baboon. Intracerebroventricular infusions of CRF or Ucn significantly decreased daily food intake. The decrease with Ucn continued into the postinfusion period. These infusions did not alter daily water intake. Daily voluntary intake of 300 mM NaCl solution was not increased, and there was evidence of reductions on days 2-4 of the infusions. Intramuscular injections of porcine ACTH or synthetic ACTH (Synacthen) for 5 days did not affect daily NaCl intake, although the doses were sufficient to increase cortisol secretion and arterial blood pressure. Sodium depletion by 3 days of furosemide injections did induce a characteristic sodium appetite in the same baboons. These results demonstrate the anorexigenic action of CRF and Ucn in this primate. Also, CRF, Ucn, and ACTH did not stimulate sodium appetite at the doses used.


Subject(s)
Adrenocorticotropic Hormone/pharmacology , Corticotropin-Releasing Hormone/pharmacology , Eating/physiology , Feeding Behavior/physiology , Stress, Physiological/physiopathology , Animals , Diuretics/pharmacology , Drinking/drug effects , Drinking/physiology , Eating/drug effects , Feeding Behavior/drug effects , Furosemide/pharmacology , Hypothalamus/physiology , Injections, Intramuscular , Injections, Intraventricular , Male , Papio , Sodium/deficiency , Sodium, Dietary/pharmacology , Urocortins
13.
J Biol Chem ; 276(34): 31528-34, 2001 Aug 24.
Article in English | MEDLINE | ID: mdl-11425856

ABSTRACT

The first extracellular domain (ECD-1) of the corticotropin releasing factor (CRF) type 1 receptor, (CRFR1), is important for binding of CRF ligands. A soluble protein, mNT-CRFR1, produced by COS M6 cells transfected with a cDNA encoding amino acids 1--119 of human CRFR1 and modified to include epitope tags, binds a CRF antagonist, astressin, in a radioreceptor assay using [(125)I-d-Tyr(0)]astressin. N-terminal sequencing of mNT-CRFR1 showed the absence of the first 23 amino acids of human CRFR1. This result suggests that the CRFR1 protein is processed to cleave a putative signal peptide corresponding to amino acids 1--23. A cDNA encoding amino acids 24--119 followed by a FLAG tag, was expressed as a thioredoxin fusion protein in Escherichia coli. Following thrombin cleavage, the purified protein (bNT-CRFR1) binds astressin and the agonist urocortin with high affinity. Reduced, alkylated bNT-CRFR1 does not bind [(125)I-D-Tyr(0)]astressin. Mass spectrometric analysis of photoaffinity labeled bNT-CRFR1 yielded a 1:1 complex with ligand. Analysis of the disulfide arrangement of bNT-CRFR1 revealed bonds between Cys(30) and Cys(54), Cys(44) and Cys(87), and Cys(68) and Cys(102). This arrangement is similar to that of the ECD-1 of the parathyroid hormone receptor (PTHR), suggesting a conserved structural motif in the N-terminal domain of this family of receptors.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Amino Acid Sequence , Animals , COS Cells , Circular Dichroism , Corticotropin-Releasing Hormone/chemistry , Corticotropin-Releasing Hormone/isolation & purification , DNA, Complementary , Humans , Molecular Sequence Data , Solubility
14.
Proc Natl Acad Sci U S A ; 98(13): 7570-5, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11416224

ABSTRACT

The corticotropin-releasing factor (CRF) family of neuropeptides includes the mammalian peptides CRF, urocortin, and urocortin II, as well as piscine urotensin I and frog sauvagine. The mammalian peptides signal through two G protein-coupled receptor types to modulate endocrine, autonomic, and behavioral responses to stress, as well as a range of peripheral (cardiovascular, gastrointestinal, and immune) activities. The three previously known ligands are differentially distributed anatomically and have distinct specificities for the two major receptor types. Here we describe the characterization of an additional CRF-related peptide, urocortin III, in the human and mouse. In searching the public human genome databases we found a partial expressed sequence tagged (EST) clone with significant sequence identity to mammalian and fish urocortin-related peptides. By using primers based on the human EST sequence, a full-length human clone was isolated from genomic DNA that encodes a protein that includes a predicted putative 38-aa peptide structurally related to other known family members. With a human probe, we then cloned the mouse ortholog from a genomic library. Human and mouse urocortin III share 90% identity in the 38-aa putative mature peptide. In the peptide coding region, both human and mouse urocortin III are 76% identical to pufferfish urocortin-related peptide and more distantly related to urocortin II, CRF, and urocortin from other mammalian species. Mouse urocortin III mRNA expression is found in areas of the brain including the hypothalamus, amygdala, and brainstem, but is not evident in the cerebellum, pituitary, or cerebral cortex; it is also expressed peripherally in small intestine and skin. Urocortin III is selective for type 2 CRF receptors and thus represents another potential endogenous ligand for these receptors.


Subject(s)
Brain/metabolism , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Peptide Fragments/pharmacology , Receptors, Corticotropin-Releasing Hormone/metabolism , Amino Acid Sequence , Animals , CHO Cells , Corticotropin-Releasing Hormone/chemistry , Cricetinae , Cyclic AMP/metabolism , Genome, Human , Humans , Kinetics , Mice , Molecular Sequence Data , Organ Specificity , Peptide Fragments/chemical synthesis , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/physiology , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcription, Genetic , Transfection , Urocortins
15.
Clin Exp Pharmacol Physiol ; 28(3): 244-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11236135

ABSTRACT

1. The anterior pituitary is well documented to be under the control of central and peripheral factors that dynamically interact to affect cell-specific modulation of pituitary functions. However, it is becoming increasingly evident that these extrinsic factors work in concert with a variety of local products that exert autocrine/paracrine control on pituitary cells. 2. These factors modulate the activity of their target pituitary cells by altering the synthesis and secretion of cell-specific hormones and by exerting control on the growth and differentiation of cells of this tissue. Included in the list of growth factors and bioactive peptides known to be products of pituitary cells are the activins, possibly inhibins and follistatins. 3. These protein factors play an important role in the local modulation of several pituitary cell types and are crucial for the maintenance of normal follicle-stimulating hormone production and, thus, reproductive function and fertility.


Subject(s)
Glycoproteins/physiology , Growth Substances/physiology , Inhibins/physiology , Activins , Animals , Follistatin , Glycoproteins/pharmacology , Growth Substances/pharmacology , Humans , Inhibins/antagonists & inhibitors , Pituitary Gland/metabolism , Pituitary Gland/physiology
16.
Proc Natl Acad Sci U S A ; 98(5): 2843-8, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11226328

ABSTRACT

Here we describe the cloning and initial characterization of a previously unidentified CRF-related neuropeptide, urocortin II (Ucn II). Searches of the public human genome database identified a region with significant sequence homology to the CRF neuropeptide family. By using homologous primers deduced from the human sequence, a mouse cDNA was isolated from whole brain poly(A)(+) RNA that encodes a predicted 38-aa peptide, structurally related to the other known mammalian family members, CRF and Ucn. Ucn II binds selectively to the type 2 CRF receptor (CRF-R2), with no appreciable activity on CRF-R1. Transcripts encoding Ucn II are expressed in discrete regions of the rodent central nervous system, including stress-related cell groups in the hypothalamus (paraventricular and arcuate nuclei) and brainstem (locus coeruleus). Central administration of 1-10 microg of peptide elicits activational responses (Fos induction) preferentially within a core circuitry subserving autonomic and neuroendocrine regulation, but whose overall pattern does not broadly mimic the CRF-R2 distribution. Behaviorally, central Ucn II attenuates nighttime feeding, with a time course distinct from that seen in response to CRF. In contrast to CRF, however, central Ucn II failed to increase gross motor activity. These findings identify Ucn II as a new member of the CRF family of neuropeptides, which is expressed centrally and binds selectively to CRF-R2. Initial functional studies are consistent with Ucn II involvement in central autonomic and appetitive control, but not in generalized behavioral activation.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Amino Acid Sequence , Animals , Behavior, Animal , CHO Cells , Cloning, Molecular , Corticotropin-Releasing Hormone/chemistry , Corticotropin-Releasing Hormone/genetics , Cricetinae , Male , Molecular Sequence Data , Polymerase Chain Reaction , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/metabolism , Sequence Homology, Amino Acid , Urocortins
17.
Endocrinology ; 142(3): 1065-72, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181520

ABSTRACT

Activins and transforming growth factor-beta (TGF beta) are crucial autocrine, paracrine, and endocrine modulators of anterior pituitary function. Activins regulate most pituitary cells and lactotropes are targets of TGF beta. Smad2 and Smad3 are two cellular mediators of activin/TGF beta signaling, whereas Smad7 is as an inducible, negative modulator of the pathway. This study was undertaken to evaluate Smad7 regulation in the pituitary. Activin A rapidly and transiently increased Smad7 messenger RNA (mRNA) levels of rat anterior pituitary (RAP), clonal gonadotrope (alpha T 3-1 and L beta T2), and corticotrope (AtT20) cells with an EC(50) of 0.1-0.2 nM. In RAP cells, activin A or TGF beta 1 had equivalent effects that were additive. Follistatin, known to bind and inactivate activins, prevented Smad7 induction by activin. Inhibin A partially antagonized activin A, perhaps reflecting gonadotrope-selective actions. This antagonism was also evident with alpha T 3-1 and L beta T2 gonadotropes. Forskolin had no measurable effect in RAP cells, but increased Smad7 mRNA levels in alpha T3-1 cells and decreased them in L beta T2 cells. Transient transfection of Smad7 along with 3TPLux, an activin/TGF beta-responsive reporter, blocked activin-mediated promoter activation in alpha T3-1 and AtT20 cells. In alpha T3-1 cells, which express endogenous follistatin mRNA, a follistatin-luciferase reporter, rFS(rin3)-Luc, was transcriptionally activated by activin A, or when cotransfected with a constitutively active ActRIB [Alk4(T>D)], Smad2, or Smad3. Smad7 blocked rFS(rin3)-Luc activation by activin A or Alk4(T>D). Together, these results point to a role of Smad7 in modulating activin/TGF beta signaling in the pituitary.


Subject(s)
DNA-Binding Proteins/physiology , Inhibins/physiology , Pituitary Gland, Anterior/physiology , Signal Transduction/physiology , Trans-Activators/physiology , Transforming Growth Factor beta/physiology , Activins , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Follistatin , Glycoproteins/pharmacology , Inhibins/pharmacology , Male , Mice , Osmolar Concentration , Pituitary Gland, Anterior/cytology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Smad7 Protein , Time Factors , Trans-Activators/genetics , Transcription, Genetic , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1
18.
Neuroscience ; 101(1): 115-29, 2000.
Article in English | MEDLINE | ID: mdl-11068141

ABSTRACT

The corticotropin-releasing factor-binding protein is distinct from known corticotropin-releasing factor receptors, but can bind the peptide and neutralize its biological actions. Recent interest has centered about the therapeutic potential of "ligand inhibitors" of binding protein action, synthetic corticotropin-releasing factor fragments which are inactive at corticotropin-releasing factor receptors, but can displace the peptide from the binding protein, thereby increasing levels of free corticotropin-releasing factor. To identify sites of action of such ligands, the distribution of Fos expression seen following intracerebroventricular administration of rat/human corticotropin-releasing factor(6-33) (5-50 microg) was charted in relation to corticotropin-releasing factor-binding protein and receptor expression. It was expected that Fos induction would mimic aspects of the distribution of the two known corticotropin-releasing factor receptors, but the far greater correspondence was seen with that of the binding protein itself. This included neurons in the isocortex, the olfactory system, amygdala and a number of discrete brainstem cell groups; many Fos-immunoreactive neurons in each were found to co-express corticotropin-releasing factor-binding protein messenger RNA. Subsets of activated neurons co-expressed Type 1 corticotropin-releasing factor receptor messenger RNA, though these were largely limited to cell groups that also express the corticotropin-releasing factor-binding protein, and where binding protein immunoreactivity and Type 1 receptor transcripts were found to co-exist. Responsive neurons displaying Type 2 corticotropin-releasing factor receptor message were seen reliably only in the lateral septal nucleus. These findings support only a limited capacity of the ligand inhibitor to activate neurons bearing corticotropin-releasing factor receptors. The more pervasive activation seen among neurons that express the corticotropin-releasing factor-binding protein may be indicative of an unexpected role for this protein in signaling by corticotropin-releasing factor-related peptides.


Subject(s)
Brain/drug effects , Carrier Proteins/drug effects , Corticotropin-Releasing Hormone/pharmacology , Neurons/drug effects , Peptide Fragments/pharmacology , Animals , Brain/cytology , Brain/metabolism , Carrier Proteins/metabolism , Corticotropin-Releasing Hormone/drug effects , Corticotropin-Releasing Hormone/metabolism , Dose-Response Relationship, Drug , Ligands , Male , Neurons/cytology , Neurons/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/drug effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Time Factors , Up-Regulation/drug effects , Up-Regulation/physiology , Urocortins
19.
Proc Natl Acad Sci U S A ; 97(18): 10277-81, 2000 Aug 29.
Article in English | MEDLINE | ID: mdl-10963687

ABSTRACT

Constitutive activity, or ligand-independent activity, of mutant G protein-coupled receptors (GPCRs) has been described extensively and implicated in the pathology of many diseases. Using the corticotropin-releasing factor (CRF) receptor and the thrombin receptor as a model, we present a ligand-dependent constitutive activation of a GPCR. A chimera in which the N-terminal domain of the CRF receptor is replaced by the amino-terminal 16 residues of CRF displays significant levels of constitutive activation. The activity, as measured by intracellular levels of cAMP, is blocked in a dose-dependent manner by the nonpeptide antagonist antalarmin. These results support a propinquity effect in CRF receptor activation, in which the amino-terminal portion of the CRF peptide is presented to the body of the receptor in the proper proximity for activation. This form of ligand-dependent constitutive activation may be of general applicability for the creation of constitutively activated GPCRs that are regulated by peptide ligands such as CRF. These chimeras may prove useful in analyzing mechanisms of receptor regulation and in the structural analysis of ligand activated receptors.


Subject(s)
Receptors, Corticotropin-Releasing Hormone/physiology , Receptors, Thrombin/physiology , Amino Acid Sequence , Animals , COS Cells , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/pharmacology , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Structure, Secondary , Receptors, Corticotropin-Releasing Hormone/chemistry , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Thrombin/chemistry , Receptors, Thrombin/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transfection , Urocortins
20.
Endocrinology ; 141(8): 2715-24, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10919255

ABSTRACT

Intracerebroventricular injection of CRF or urocortin (Ucn) reduces appetite and body weight. CRFR1 and CRFR2, the receptors for CRF and Ucn, are expressed in neurons associated with appetite-control and metabolism, but their relative contributions in mediating CRF- or Ucn-induced hypophagia and weight loss are not known. We used homozygous mice lacking CRFR1 (CRFR1-/-) and wild-type littermates to determine the role of CRFR1 in mediating the changes in food intake and body weight following intracerebroventricular administration of Ucn. CRFR1-/- mice, which are glucocorticoid deficient, were given corticosterone in their drinking water to induce diurnal variations in circulating corticosterone. A 7-day intracerebroventricular infusion of Ucn transiently suppressed ad libitum food intake equally in CRFR1-/- and wild-type mice. Body weight reduction during Ucn infusion paralleled food intake in wild-type mice, but persisted throughout the infusion in CRFR1-/- mice. After food-deprivation, acute intracerebroventricular injection of Ucn suppressed food intake for 1.5 h in wild-type mice. By contrast, CRFR1-/- mice did not respond to Ucn 1.5 h after injection. At later time points, Ucn suppressed food intake equally in both genotypes. The distinct time courses of CRF-receptor-induced hypophagia suggest that separate pathways act cooperatively to adjust food intake during challenges to homeostasis.


Subject(s)
Appetite/physiology , Corticotropin-Releasing Hormone/pharmacology , Eating/physiology , Receptors, Corticotropin-Releasing Hormone/deficiency , Receptors, Corticotropin-Releasing Hormone/physiology , Weight Loss/drug effects , Adrenocorticotropic Hormone/pharmacology , Animals , Appetite/drug effects , Circadian Rhythm , Corticosterone/administration & dosage , Corticosterone/blood , Corticotropin-Releasing Hormone/administration & dosage , Drinking , Eating/drug effects , Food Deprivation , Injections, Intraventricular , Male , Mice , Mice, Knockout , Receptors, Corticotropin-Releasing Hormone/genetics , Urocortins
SELECTION OF CITATIONS
SEARCH DETAIL
...