Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 207(2): 353-9, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-19840820

ABSTRACT

Conditioned opiate reward (COR) is rapidly acquired and slowly extinguished. The slow rate of extinction of the salience of drug-related cues contributes to drug craving and relapse. The gamma-aminobutyric acid receptor type B (GABA(B)) agonist, baclofen, attenuates the unconditioned rewarding actions of several drugs of abuse and was investigated for effects on the extinction of COR. C57BL/6 mice were utilized in an unbiased conditioned place preference (CPP) protocol using morphine (10mg/kg, s.c.) and saline. CPP was measured by increases in time spent in the morphine-associated (CS+) vs. the saline-associated (CS-) chamber in a 15-min test after four morphine and four saline alternated conditioning sessions. CPP and locomotor sensitization were produced to the CS+ chamber. Subsequently, sixteen daily extinction sessions were conducted with either vehicle or baclofen (1 or 2.5mg/kg, s.c.) treatment given either before or after the sessions. This design was used to create the baclofen drug state before or after the activation of the CPP memory trace in the extinction protocol. After morphine CPP development, its extinction was significantly facilitated in a dose-dependent manner by post-session, but not by pre-session, baclofen treatments. No significant sedative effects of baclofen were detected during any extinction training or testing phase. Baclofen treatment facilitated the extinction of COR and reduced conditioned sensitization during extinction when given after, but not before, the activation of the CPP memory trace. Baclofen appears to have disrupted reconsolidation of conditioned reward memory during extinction training and might similarly facilitate extinction learning in human opiate addiction.


Subject(s)
Analgesics, Opioid/pharmacology , Baclofen/pharmacology , Conditioning, Classical/drug effects , Extinction, Psychological/drug effects , GABA Agonists/pharmacology , Morphine/pharmacology , Animals , Baclofen/administration & dosage , Dose-Response Relationship, Drug , GABA Agonists/administration & dosage , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Space Perception/drug effects , Time Factors
2.
Neuroscience ; 145(1): 33-41, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17207935

ABSTRACT

The EL/Suz (EL) mouse is a model of multifactorial temporal lobe epilepsy in which seizures begin around 90 days of age, but can be hastened through increased exposure to human handling. In order to better understand seizure etiology in this mouse strain relative to seizure-resistant control mice, the present study examined region-specific neuronal activation in response to non-seizure-inducing handling implemented before the onset of seizure susceptibility. Immediate-early gene (cFos) expression emerged in EL mice by postnatal day (PND) 21 in the primary motor cortex, progressed to the locus ceruleus and prefrontal cortex by PND 35, and appeared in the hippocampus and amygdala by PND 70, as mice neared the age of onset for seizure susceptibility. Thus, mirroring the pattern observed during the course of a seizure, specific brain regions were differentially recruited to a neural network for seizure predisposition before the onset of seizure susceptibility. This developmental pattern of early and transient neural activation represents an important window for the study of causal mechanisms of seizure susceptibility following exposure to environmental triggers.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Genetic Predisposition to Disease , Nerve Net/physiopathology , Seizures , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Disease Models, Animal , Female , Hindlimb Suspension/methods , Immunohistochemistry/methods , Mice , Mice, Neurologic Mutants , Models, Biological , Motor Activity/physiology , Nerve Net/pathology , Proto-Oncogene Proteins c-fos/metabolism , Seizures/genetics , Seizures/pathology , Seizures/physiopathology
3.
Behav Brain Res ; 125(1-2): 81-8, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11682097

ABSTRACT

Ingestive behaviors, feeding and drinking, constitute unconditioned, obligatory functions that are tightly regulated in the rodent according to demands of the external and internal milieu. Dependent measures of food intake have been used extensively in rats to infer the identity and function of neurochemical pathways, which mediate energy balance. A recent interest in application of appetitive measures in mice can be attributed jointly to the discovery of novel markers of energy balance in genetically obese mice as well as systematic targeting of known feeding regulatory pathways in bioengineered mutant mice. Accordingly, this review will attempt to provide the reader interested in behavioral phenotyping of knockout or transgenic mice with information regarding the ethology of mouse eating behavior, known mechanisms of appetitive regulation and examples of successes and pitfalls encountered when studying food intake in mutant mice.


Subject(s)
Energy Metabolism/genetics , Ethology , Feeding Behavior/physiology , Mice, Mutant Strains/genetics , Phenotype , Animals , Mice , Mice, Knockout/genetics , Mice, Obese/genetics , Mice, Transgenic/genetics , Rats , Species Specificity
4.
Physiol Behav ; 74(1-2): 5-13, 2001.
Article in English | MEDLINE | ID: mdl-11564446

ABSTRACT

Acute administration of corticotropin-releasing factor (CRF) results in anorexic and sympathomimetic effects that suggest efficacy in chronic models of energy balance. The present studies employed a broad spectrum energy balance indices in lean and genetically obese Zucker rats in order to fully characterize the pharmacological efficacy of CRF and a CRF binding protein (CRF-BP) ligand inhibitor, CRF(6-33), which is thought to liberate CRF from CRF-BP. Acute administration of CRF(6-33) significantly increased CRF(2) receptor density by 10% within the ventromedial hypothalamic (VMH) nucleus of Zucker lean rats and decreased density by 10% in Zucker obese rats. A single infusion of CRF(6-33) increased nonshivering thermogenesis by 25-30% as measured by proton conductance in brown adipose tissue of both lean and obese rats. Chronic CRF(6-33) infusion suppressed body weight gain and elevated core temperature irrespective of genotype while increasing motor activity in obese rats without altering heart rate or blood pressure. Taken together, these results document strain-dependent, long-term effects of a CRF-BP ligand inhibitor on a select subset of physiological and behavioral measures of increased energy expenditure.


Subject(s)
Corticotropin-Releasing Hormone/physiology , Energy Metabolism/drug effects , Adipose Tissue, Brown/drug effects , Adrenocorticotropic Hormone/metabolism , Adrenocorticotropic Hormone/pharmacology , Animals , Autoradiography , Body Temperature/drug effects , Brain Chemistry/drug effects , Brain Chemistry/physiology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Circadian Rhythm/drug effects , Corticotropin-Releasing Hormone/metabolism , Guanosine Diphosphate/metabolism , Hemodynamics/drug effects , Injections, Intraventricular , Ligands , Male , Rats , Rats, Zucker , Telemetry , Thermogenesis/drug effects , Weight Gain/drug effects
5.
Pharmacol Rev ; 53(2): 209-43, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356984

ABSTRACT

The goal of this article is to summarize available data examining the physiological significance of brain corticotropin-releasing factor (CRF) systems in mediating the behavioral and physiological effects of several classes of abused drugs, including opioid and psychostimulant drugs, alcohol and sedative hypnotics, nicotine, and cannabinoids. An initial discussion of CRF neurobiology is followed by consideration of the role of CRF in drug-induced activation of the hypothalamic-pituitary-adrenocortical (HPA) axis, the behavioral effects of drugs (e.g., locomotor activity, anxiogenic-like responses), drug self-administration, drug withdrawal, and relapse to drug-seeking. Subsequently, neurochemical changes in brain CRF in response to acute and chronic drug exposure are examined. A major conclusion derived from the data reviewed is that extrahypothalamic brain CRF systems are critically involved in behavioral and physiological manifestations of drug withdrawal and in relapse to drug-taking behavior induced by environmental stressors. On the other hand, it appears that hypothalamic CRF, via its action on the HPA axis, is involved in the reinforcing effects of cocaine and alcohol, and the locomotor activating effects of psychostimulant drugs. These preclinical data may provide a rationale for the development of CRF-based pharmacotherapies for the treatment of compulsive drug use in humans.


Subject(s)
Brain/metabolism , Corticotropin-Releasing Hormone/physiology , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Substance-Related Disorders/metabolism , Animals , Brain/drug effects , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Humans , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Substance Withdrawal Syndrome/metabolism
6.
Peptides ; 22(5): 713-24, 2001 May.
Article in English | MEDLINE | ID: mdl-11337084

ABSTRACT

Corticotropin-releasing hormone (CRH) and urocortin in the central nervous system affect behavior and can enhance behavioral responses to stressors. The action of CRH-related peptides is mediated through multiple receptors that differ markedly in their pharmacological profiles and anatomical distribution. Comparative pharmacology of CRH receptor agonists suggests that CRH, urocortin, sauvagine and urotensin consistently mimic, and CRH receptor antagonists consistently lessen, functional consequences of stressor exposure. Recently, important advances have been made in understanding the CRH system and its role in behavioral responses to stress by the development of specific CRH receptor antagonists, application of antisense oligonucleotides and development of transgenic mice lacking peptides and functional receptors. This review summarizes recent findings with respect to components of the CRH system and their role in stress-induced behavioral responses.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Stress, Psychological/drug therapy , Stress, Psychological/physiopathology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Dose-Response Relationship, Drug , Humans , Oligonucleotides, Antisense/pharmacology , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Urocortins
7.
Brain Res ; 900(2): 177-85, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11334796

ABSTRACT

Corticotropin-releasing factor (CRF) receptor agonist and CRF binding-protein (CRF-BP) ligand inhibitor peptides both activate CRF systems but exert very distinct functional profiles in animal models of arousal, energy balance and emotionality. The present studies were designed to extend the dissimilar efficacy profiles of central administration of a CRF agonist, r/h CRF(1-41), versus a CRF-BP ligand inhibitor, r/h CRF(6-33), into mouse and rat models of energy balance in order to further explore in vivo efficacy of these ligands in two separate animal species. In CD-1 mice, food intake was significantly attenuated 3 h after acute administration of CRF(1-41) (0.007-0.2 nmol), but not CRF(6-33). In obese Ob/Ob mice, both CRF(1-41) (0.007-0.2 nmol) and CRF(6-33) (0.02-2.3 nmol) significantly attenuated basal feeding over 3 h following acute peptide administration. In rats, CRF(1-41) (1 nmol) and CRF(6-33) (1.5-7.7 nmol) infusion significantly increased rectal temperature. In studies employing a telemetry apparatus, core temperature was also increased by CRF(1-41) (1 nmol) and CRF(6-33) (1.5 nmol), whereas only CRF(1-41) increased locomotor activity and heart rate. These results suggest that CRF receptor agonist administration is capable of producing a global profile of negative energy balance by reducing food intake in mice and increasing energy expenditure in rats. In contrast, CRF-BP ligand inhibitor administration appears to suppress food intake in a mouse strain selective manner and to elevate rectal and core temperature in rats without accompanying cardiovascular activation.


Subject(s)
Body Temperature/drug effects , Carrier Proteins/antagonists & inhibitors , Corticotropin-Releasing Hormone/antagonists & inhibitors , Eating/drug effects , Peptide Fragments/pharmacology , Amphetamine/pharmacology , Animals , Carrier Proteins/chemistry , Corticotropin-Releasing Hormone/analogs & derivatives , Corticotropin-Releasing Hormone/chemistry , Corticotropin-Releasing Hormone/pharmacology , Humans , Male , Mice , Mice, Inbred Strains , Peptide Fragments/chemistry , Rats , Rats, Wistar , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Rectum/physiology , Sheep , Telemetry
8.
Behav Brain Res ; 122(1): 43-50, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11287075

ABSTRACT

Behavioral actions of centrally administered corticotropin-releasing factor (CRF) are likely subserved by multiple brain targets and functional effector systems. The present studies compared effects of two CRF ligands, a full, post-synaptic CRF receptor agonist (rat/human CRF(1-41)) and a CRF binding protein ligand inhibitor (rat/human CRF(6-33)) in a behavioral testing battery sensitive to arousal, fear-like and aversive processes in Wistar rats. The profile of global efficacy for the centrally administered CRF receptor agonist was characterized by low dose (0.5-1.0 microg) arousal-like effects in locomotor and conditioned ambulation contexts and by high dose (5-25 microg) conditioned immobility, taste aversion and place aversion. In contrast, a profile of limited efficacy for the centrally administered CRF binding protein ligand inhibitor included only dose dependent motor activating and facilitation of fear conditioning effects without any of the anxiogenic-like or aversive properties of CRF agonist administration. These results suggest that arousal-like activation is a fundamental, physiologically relevant consequence of brain CRF system stimulation whereas aversive and anxiety-like effects reflect pharmacological actions of a CRF receptor agonist.


Subject(s)
Arousal/drug effects , Carrier Proteins/antagonists & inhibitors , Corticotropin-Releasing Hormone/physiology , Fear/drug effects , Receptors, Corticotropin-Releasing Hormone/agonists , Animals , Avoidance Learning/drug effects , Conditioning, Psychological/drug effects , Corticotropin-Releasing Hormone/agonists , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/metabolism , Dose-Response Relationship, Drug , Ligands , Male , Rats , Rats, Wistar
9.
Expert Opin Investig Drugs ; 10(4): 647-59, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11281815

ABSTRACT

Activation of the corticotropin-releasing factor (CRF) family of neuropeptide receptors in the brain and periphery appears to mediate stress-related changes in a variety of physiological and functional domains. Comparative pharmacology of CRF receptor agonists suggests that CRF, urocortin, sauvagine and urotensin consistently mimic, and conversely peptide CRF receptor antagonists lessen, the functional consequences of stressor exposure. Together with the development of novel non-peptide CRF receptor antagonists, a growing number of CRF receptor selective ligands are available to elucidate the neurobiology and physiological role of CRF systems. The present review considers available preclinical evidence as well as results from one Phase II clinical trial which address the hypothesis that CRF receptor antagonists may represent a new option for pharmacotherapy of stress-related disorders.


Subject(s)
Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Animals , Anti-Anxiety Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Feeding and Eating Disorders/drug therapy , Gastrointestinal Motility/drug effects , Humans , Intestinal Diseases/drug therapy , Receptors, Corticotropin-Releasing Hormone/physiology
10.
Eur J Neurosci ; 14(12): 2003-10, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11860495

ABSTRACT

Pregnenolone sulphate (PREGS) has generated interest as one of the most potent memory-enhancing neurosteroids to be examined in rodent learning studies, with particular importance in the ageing process. The mechanism by which this endogenous steroid enhances memory formation is hypothesized to involve actions on glutamatergic and GABAergic systems. This hypothesis stems from findings that PREGS is a potent positive modulator of N-methyl-d-aspartate receptors (NMDARs) and a negative modulator of gamma-aminobutyric acid(A) receptors (GABA(A)Rs). Moreover, PREGS is able to reverse the amnesic-like effects of NMDAR and GABA(A)R ligands. To investigate this hypothesis, the present study in rats examined the memory-altering abilities of structural analogs of PREGS, which differ in their modulation of NMDAR and/or GABA(A)R function. The analogs tested were: 11-ketopregnenolone sulphate (an agent that is inactive at GABA(A)Rs and NMDARs), epipregnanolone ([3beta-hydroxy-5beta-pregnan-20-one] sulphate, an inhibitor of both GABA(A)Rs and NMDARs), and a newly synthesized (-) PREGS enantiomer (which is identical to PREGS in effects on GABA(A)Rs and NMDARs). The memory-enhancing effects of PREGS and its analogs were tested in the passive avoidance task using the model of scopolamine-induced amnesia. Both PREGS and its (-) enantiomer blocked the effects of scopolamine. The results show that, unlike PREGS, 11-ketopregnenolone sulphate and epipregnanolone sulphate failed to block the effect of scopolamine, suggesting that altering the modulation of NMDA receptors diminishes the memory-enhancing effects of PREGS. Moreover, enantioselectivity was demonstrated by the ability of natural PREGS to be an order of magnitude more effective than its synthetic enantiomer in reversing scopolamine-induced amnesia. These results identify a novel neuropharmacological site for the modulation of memory processes by neuroactive steroids.


Subject(s)
Amnesia/drug therapy , Avoidance Learning/drug effects , Brain/drug effects , Memory/drug effects , Pregnanediol/analogs & derivatives , Pregnenolone/analogs & derivatives , Receptors, GABA-A/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Amnesia/metabolism , Amnesia/physiopathology , Animals , Animals, Newborn , Avoidance Learning/physiology , Brain/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Memory/physiology , Muscarinic Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Pregnanediol/pharmacology , Pregnanolone/pharmacology , Pregnenolone/metabolism , Pregnenolone/pharmacology , Rats , Rats, Wistar , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Scopolamine/pharmacology , Stereoisomerism
11.
Peptides ; 21(3): 345-51, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10793215

ABSTRACT

Corticotropin-releasing hormone (CRH) is a potent regulator of the hypothalamic-pituitary-adrenal axis, and reduces food intake when administered into the third cerebral ventricle (i3vt). However, CRH also promotes conditioned taste aversion (CTA) learning which indicates that its anorectic effects are accompanied by aversive consequences that would reduce food intake independently of energy regulation. Urocortin (Ucn) is a closely related mammalian peptide that binds to both identified CRH receptor subtypes and also reduces food intake when administered i3vt. The present experiments compared the aversive consequences of i3vt administration of CRH and Ucn at doses that produced comparable decrements in food intake. Experiment 1 found that 1.0 microg Ucn and 2.0 microg CRH produced similar reductions in food intake. Experiment 2 demonstrated that, at these doses, CRH but not Ucn promoted robust and reliable CTA learning. A third experiment showed comparable increased c-Fos-like immunoreactivity after Ucn and CRH in forebrain and hindbrain structures associated with food intake. It is concluded that Ucn, at doses that reduce food intake to levels like that observed after administration of CRH, do not produce similarly aversive consequences.


Subject(s)
Avoidance Learning/drug effects , Cerebral Ventricles/physiology , Corticotropin-Releasing Hormone/pharmacology , Energy Intake/drug effects , Genes, fos , Taste , Amygdala/drug effects , Amygdala/metabolism , Animals , Cerebral Ventricles/drug effects , Corticotropin-Releasing Hormone/administration & dosage , Feeding Behavior/drug effects , Gene Expression Regulation/drug effects , Injections, Intraventricular , Male , Rats , Rats, Long-Evans , Reaction Time , Saccharin , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism , Urocortins
12.
Endocrinology ; 141(2): 498-504, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10650928

ABSTRACT

CRF is the main component in the brain neuropeptide effector system responsible for the behavioral, endocrine, and physiological activation that accompanies stress activation. Reduced CRF system activation plays a role in the etiology of a variety of psychiatric and metabolic disease states. We have developed a novel protein conjugate that joins native rat/human CRF to a ribosome-inactivating protein, saporin (CRF-SAP), for the purpose of targeted inactivation of CRF receptor-expressing cells. Cytotoxicity measurements revealed that CRF-SAP (1-100 nM) produced concentration-dependent and progressive cell death over time in CRF1 receptor-transfected L cells, but at similar concentrations had no effect on CRF2alpha receptor-transfected cells. The CRF-SAP-induced toxicity in CRF1-transfected cells was prevented by coincubation with the competitive CRF1/CRF2 receptor peptide antagonist, [D-Phe12]CRF-(12-41), or the selective nonpeptide CRF1 receptor antagonist, NBI 27914. Finally, in cultured rat pituitary cells that express native CRF1 receptors, CRF-SAP suppressed CRF-induced (1 nM) ACTH release. GnRH (1-10 nM) stimulated LH release was also assessed in the same pituitary cultures. Although there was a slight decrease in LH release from these cultures, this decrease was observed with CRF-SAP or SAP alone, suggesting that the response was nonspecific. Taken together, these results suggest the utility of CRF-SAP as a specific and subtype-selective tool for long term impairment of CRF1 receptor-expressing cells.


Subject(s)
Corticotropin-Releasing Hormone/pharmacology , N-Glycosyl Hydrolases , Plant Proteins/pharmacology , Receptors, Corticotropin-Releasing Hormone/physiology , Adrenocorticotropic Hormone/metabolism , Animals , Carrier Proteins/metabolism , Cell Survival/drug effects , Cells, Cultured , Female , Gonadotropin-Releasing Hormone/pharmacology , Humans , Immunotoxins/pharmacology , L Cells , Luteinizing Hormone/metabolism , Mice , Pituitary Gland/cytology , Pituitary Gland/drug effects , Pituitary Gland/physiology , Rats , Receptors, Corticotropin-Releasing Hormone/drug effects , Receptors, Corticotropin-Releasing Hormone/genetics , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Ribosome Inactivating Proteins, Type 1 , Saporins , Transfection
13.
Brain Res ; 848(1-2): 141-52, 1999 Nov 27.
Article in English | MEDLINE | ID: mdl-10612706

ABSTRACT

Corticotropin-releasing factor (CRF) and CRF-related neuropeptides have an important role in the central nervous system to mediate behavioral responses to stressors. CRF receptor antagonists are very effective in reversing stress-induced suppression and activation in behavior. An additional CRF-like neuropeptide, urocortin, has been identified in the brain and has a high affinity for the CRF-2 receptor in addition to the CRF-1 receptor. Urocortin has many of the effects of CRF but also is significantly more potent than CRF in decreasing feeding in both meal-deprived and free-feeding rats. In mouse genetic models, mice over-expressing CRF show anxiogenic-like responses compared to wild-type mice, and mice lacking the CRF-1 receptor showed an anxiolytic-like behavioral profile compared to wild-type mice. Results to date have led to the hypothesis that CRF-1 receptors may mediate CRF-like neuropeptide effects on behavioral responses to stressors, but CRF-2 receptors may mediate the suppression of feeding produced by CRF-like neuropeptides. Brain sites for the behavioral effects of CRF include the locus coeruleus (LC), paraventricular nucleus (PVN) of the hypothalamus, the bed nucleus of the stria terminalis (BNST), and the central nucleus of the amygdala. CRF may also be activated during acute withdrawal from all major drugs of abuse, and recent data suggest that CRF may contribute to the dependence and vulnerability to relapse associated with chronic administration of drugs of abuse. These data suggest that CRF systems in the brain have a unique role in mediating behavioral responses to diverse stressors. These systems may be particularly important in situations were an organism must mobilize not only the pituitary adrenal system, but also the central nervous system in response to environmental challenge. Clearly, dysfunction in such a fundamental brain-activating system may be the key to a variety of pathophysiological conditions involving abnormal responses to stressors such as anxiety disorders, affective disorders, and anorexia nervosa.


Subject(s)
Behavior, Animal/physiology , Corticotropin-Releasing Hormone/physiology , Stress, Physiological/physiopathology , Animals , Brain Chemistry/physiology , Humans , Substance-Related Disorders/physiopathology , Urocortins
15.
Curr Pharm Des ; 5(5): 289-315, 1999 May.
Article in English | MEDLINE | ID: mdl-10213797

ABSTRACT

Corticotropin-releasing factor (CRF) has been widely implicated as playing a major role in modulating the endocrine, autonomic, behavioral and immune responses to stress. The recent cloning of multiple receptors for CRF as well as the discovery of non-peptide receptor antagonists for CRF receptors have begun a new era of CRF study. Presently, there are five distinct targets for CRF with unique cDNA sequences, pharmacology and localization. These fall into three distinct classes, encoded by three different genes and have been termed the CRF1 and CRF2 receptors (belonging to the superfamily of G-protein coupled receptors) and the CRF-binding protein. The CRF2 receptor exists as three splice variants of the same gene and have been designated CRF2a CRF2b and CRF2g. The pharmacology and localization of all of these proteins in brain has been well established. The CRF1 receptor subtype is localized primarily to cortical and cerebellar regions while the CRF2a receptor is localized to subcortical regions including the lateral septum, and paraventricular and ventromedial nuclei of the hypothalamus. The CRF2b receptor is primarily localized to heart, skeletal muscle and in the brain, to cerebral arterioles and choroid plexus. The CRF2g receptor has most recently been identified in human amygdala. Expression of these receptors in mammalian cell lines has made possible the identification of non-peptide, high affinity, selective receptor antagonists. While the natural mammalian ligands oCRF and r/hCRF have high affinity for the CRF1 receptor subtype, they have lower affinity for the CRF2 receptor family making them ineffective labels for CRF2 receptors. [125I]Sauvagine has been characterized as a high affinity ligand for both the CRF1 and the CRF2 receptor subtypes and has been used in both radioligand binding and receptor autoradiographic studies as a tool to aid in the discovery of selective small molecule receptor antagonists. A number of non-peptide CRF1 receptor antagonists that can specifically and selectively block the CRF1 receptor subtype have recently been identified. Compounds such as CP 154,526 (12), NBI 27914 (129) and Antalarmin (154) inhibit CRF-stimulation of cAMP or CRF-stimulated ACTH release from cultured rat anterior pituitary cells. Furthermore, when administered peripherally, these compounds compete for ex vivo [125I]sauvagine binding to CRF1 receptors in brain sections demonstrating their ability to cross the blood-brain-barrier. In in vivo studies, peripheral administration of these compounds attenuate stress-induced elevations in plasma ACTH levels in rats demonstrating that CRF1 receptors can be blocked in the periphery. Furthermore, peripherally administered CRF1 receptor antagonists have also been demonstrated to inhibit CRF-induced seizure activity. These data clearly demonstrate that non-peptide CRF1 receptor antagonists, when administered systemically, can specifically block central CRF1 receptors and provide tools that can be used to determine the role of CRF1 receptors in various neuropsychiatric and neurodegenerative disorders. In addition, these molecules will prove useful in the discovery and development of potential orally active therapeutics for these disorders.


Subject(s)
Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Animals , Anxiety/drug therapy , Depression/drug therapy , Drug Design , Humans , Kinetics , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptors, Corticotropin-Releasing Hormone/classification , Receptors, Corticotropin-Releasing Hormone/metabolism , Seizures/drug therapy , Substance-Related Disorders/drug therapy
16.
Article in English | MEDLINE | ID: mdl-10903813

ABSTRACT

Corticotrophin-releasing factor (CRF; interchangeable with corticotrophin-releasing hormone, CRH) is a neurohormone family of peptides which implements endocrine, physiological and behavioural responses to stressor exposure. Built-in biological diversity and selectivity of CRF system function is provided by multiple endogenous ligands and receptors which are heterogeneously distributed in both brain and peripheral tissues across species. At present, there are at least five distinct targets for CRF with unique cDNA sequences, pharmacology and localization. These fall into three distinct classes, encoded by three different genes and have been termed the CRF1 and CRF2 receptors and the CRF-binding protein. Significant gains in knowledge about the physiological role of CRF binding sites in brain have emerged recently due to the proliferation of novel, high-affinity, receptor-selective pharmacological tools as well as multiple knock-out and knock-in mutant mouse models. These results support a role for CRF binding sites in co-ordinating stress reactivity, emotionality and energy balance over the life-span of the organism.


Subject(s)
Carrier Proteins/metabolism , Central Nervous System Diseases/metabolism , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Binding Sites/physiology , Brain/metabolism , Humans
17.
Neuropeptides ; 33(1): 1-12, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10657465

ABSTRACT

Mice with transgenic expression or deletion of the CRF peptide, transgenic expression of the CRF-BP or deletion of specific CRF receptor subtypes exist and will be valuable for examining candidate mediators in animal model systems recapitulating a variety of normal function. In particular, results described in this review implicate CRF in acute emotional responses studied in animal models of anxiety and drug abstinence. CRF also appears to play a role in behavioral and physiological plasticity judging by alterations in HPA reactivity to stress, information processing and energy balance regulation in CRF mutant models. Accordingly, the creation of genetically engineered mice now permits the evaluation of contributory roles for several CRF-related gene products in the pathophysiology of a variety of complex behavioral disorders. For example, the postulated causal linkage between overactivation of CRF systems and the hyper-emotionality which characterizes human affective disorders can now be more thoroughly evaluated by examining the phenotype of CRF mutant mice in animal models of depression, dementia and substance abuse.


Subject(s)
Brain Chemistry/genetics , Corticotropin-Releasing Hormone/physiology , Mice, Knockout , Mice, Mutant Strains , Animals , Mice
18.
Neuropeptides ; 33(5): 350-9, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10657512

ABSTRACT

Participation of the hypothalamo-pituitary-adrenocortical axis, and its primary brain trigger, corticotropin-releasing factor (CRF) in the control of ingestive behavior can be inferred from data suggesting that CRF and its homologue urocortin act in brain to limit appetite following administration in rodents. Moreover, levels of endogenous CRF, CRF(1)and CRF(2)receptors and CRF-binding protein, which sequesters CRF and urocortin, are altered by changes in nutritional status brought about by food restriction/repletion. Mediation of the anorexic effects of CRF and urocortin appear not to privilege CRF(1)receptors, unlike the anxiogenic effects of CRF which are primarily a consequence of CRF(1)receptor activation. Such fear-like consequences of CRF system activation constitute a non-specific mechanism whereby the emergence of behaviors incompatible with food intake may appear to suppress appetite without affecting hunger per se. However, enhanced appetite following administration of CRF receptor antagonists and the involvement of CRF systems in sexual appetite and drug-seeking behavior all suggest a role for CRF in ingestive behavior. In particular, available evidence suggests that physiologically relevant suppression of appetite may accompany CRF system activation occurring as a consequence of stressor exposure induced by nutrient imbalance, for example, or under conditions of excessive intake or consumption of unfamiliar foodstuffs.


Subject(s)
Consummatory Behavior/physiology , Corticotropin-Releasing Hormone/physiology , Drinking/physiology , Eating/physiology , Animals , Humans , Urocortins
19.
Curr Opin Drug Discov Devel ; 2(5): 491-6, 1999 Sep.
Article in English | MEDLINE | ID: mdl-19649976

ABSTRACT

Corticotropin-releasing factor (CRF) is a pluripotent neurohormone system, which implements endocrine, physiological and behavioral responses to stressor exposure. Built-in biological diversity and selectivity of CRF system function is provided by multiple endogenous ligands and receptors, which are heterologously distributed in both brain and peripheral tissues across species. Significant gains in knowledge about the physiological role of CRF receptors in the brain have emerged over the past year due to the proliferation of novel, high affinity, receptor selective pharmacological tools as well as multiple knockout and knockin mutant mouse models. These results support a role for CRF binding sites in coordinating stress reactivity, emotionality and energy balance over the life span of the organism.

20.
Psychopharmacology (Berl) ; 136(3): 247-55, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9566810

ABSTRACT

The behavioral profile of corticotropin-releasing factor (CRF) in mediating anxiogenic-like and aversive responses to stressors may be particularly relevant for dependence and withdrawal in drug-experienced organisms. Moreover, stressful aspects of drug exposure in the drug naive organism may also induce CRF system activation. In the present studies, the dependence of aversive properties of cocaine on activation of endogenous CRF systems has been evaluated in rats using taste conditioning and runway self-administration paradigms. Systemic cocaine administration (20 mg/kg i.p.) produced a conditioned saccharin aversion which was dose-dependently potentiated by central administration of the CRF receptor antagonist, D-phe CRF (12 41). In addition, i.v. cocaine administration (0.75 mg/kg per injection i.v.) produced runway goal-box avoidance and conditioned place avoidance responses which were significantly accelerated by CRF antagonist treatment. In contrast, CRF receptor stimulation using CRF itself abolished cocaine-induced increases in goal latency in the runway paradigm. This generalized involvement of CRF systems in cocaine-related motivational/associative states is consistent with the comprehensive role of CRF in mediating emotional responses to non-drug stressors.


Subject(s)
Avoidance Learning/drug effects , Cocaine/pharmacology , Narcotics/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Animals , Corticotropin-Releasing Hormone/analogs & derivatives , Corticotropin-Releasing Hormone/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Injections, Intravenous , Injections, Intraventricular , Jugular Veins , Male , Peptide Fragments/pharmacology , Rats , Rats, Wistar , Taste/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...