Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Psychopharmacology (Berl) ; 239(8): 2407-2419, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35396673

ABSTRACT

RATIONALE: Alongside a pathological, excessive, motivation for substances of abuse, substance use disorder (SUD) patients often show a dramatic loss of interest for naturally rewarding activities, such as positive peer social interaction and food intake. Yet, pre-clinical evidence of the latter SUD features remains scarce and inconsistent. OBJECTIVES: In the current study, we investigated the effect of non-rewarding and rewarding doses of morphine upon social behaviour, motivation for and intake of palatable food, in male and female C57BL/6J mice. METHODS: First, the rewarding effects of two relatively low morphine doses (1.25 and 2.5 mg/kg) were assessed using a newly established single substance administration/conditioning trial conditioned place preference (CPP) paradigm. Then, morphine (1.25 and 2.5 mg/kg) effects upon social behaviour, motivation for and intake of palatable food were examined by the three-chamber (3-CH), an operant behaviour and a palatable food preference test, respectively. RESULTS: Morphine (2.5 mg/kg) induced CPP in both male and female mice, whereas morphine (1.25 mg/kg) induced CPP only in female mice. Both morphine doses (1.25 and 2.5 mg/kg) reduced sociability, motivation for and intake of palatable food in male and female mice, independently of cognitive function or locomotor activity. CONCLUSIONS: Female mice were more sensitive than male mice to the rewarding effects of morphine. Moreover, both a non-rewarding and a rewarding dose of morphine impaired the interest for naturally rewarding activities, indicating that brain reward systems might be more sensitive to the deleterious than to the rewarding effects of substances of abuse.


Subject(s)
Morphine , Reward , Animals , Brain , Conditioning, Operant , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred C57BL , Morphine/pharmacology , Motivation
2.
J Neurosci Res ; 100(1): 309-321, 2022 01.
Article in English | MEDLINE | ID: mdl-32725663

ABSTRACT

Poor sociability and aggressive behavior are key clinical features of opioid use disorders. The corticotropin-releasing factor (CRF) system may mediate behavioral effects of substances of abuse but its implication in substance-induced social behavior deficits and outward-directed hostility remains largely unknown. CRF signaling is mediated by two receptor types, termed CRF1 and CRF2 . The present study aimed at understanding the role for the CRF1 receptor in social and aggressive behavior induced by withdrawal from repeated opiate administration. Thus, wild-type (CRF1 +/+), CRF1 receptor heterozygous (CRF1 +/-), and null mutant (CRF1 -/-) female and male mice were treated with saline or escalating doses of morphine (20-100 mg/kg, i.p.) during six consecutive days and tested in the three-chamber task for sociability (i.e., preference for an unfamiliar same-sex conspecific vs. an object) 7 days after the last administration. Moreover, aggressive biting behavior toward the unfamiliar conspecific was assessed during the three-chamber test. Opiate withdrawal disrupted sociability in CRF1 +/+ and CRF1 +/-, but not in CRF1 -/-, female mice, without affecting aggressive biting behavior in any genotype. In contrast, opiate withdrawal did not affect sociability but increased aggressive biting behavior in male mice, independently of CRF1 receptor-deficiency. Nevertheless, in opiate-withdrawn CRF1 +/+, but not CRF1 +/- and CRF1 -/-, male mice, sociability directly correlated with aggressive biting behavior, suggesting a role for the CRF1 receptor in hostility-linked social approach. These findings demonstrate the implication of the CRF1 receptor in social behavior deficits associated with repeated opiate administration and withdrawal, revealing a new potential target for the treatment of opioid use disorders.


Subject(s)
Opiate Alkaloids , Receptors, Corticotropin-Releasing Hormone , Social Behavior , Substance Withdrawal Syndrome , Animals , Corticotropin-Releasing Hormone , Female , Male , Mice , Mice, Knockout , Receptors, Corticotropin-Releasing Hormone/genetics
3.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34083436

ABSTRACT

Prefrontal control of cognitive functions critically depends upon glutamatergic transmission and N-methyl D-aspartate (NMDA) receptors, the activity of which is regulated by dopamine. Yet whether the NMDA receptor coagonist d-serine is implicated in the dopamine-glutamate dialogue in the prefrontal cortex (PFC) and other brain areas remains unexplored. Here, using electrophysiological recordings, we show that d-serine is required for the fine-tuning of glutamatergic neurotransmission, neuronal excitability, and synaptic plasticity in the PFC through the actions of dopamine at D1 and D3 receptors. Using in vivo microdialysis, we show that D1 and D3 receptors exert a respective facilitatory and inhibitory influence on extracellular levels and activity of d-serine in the PFC, with actions expressed primarily via the cAMP/protein kinase A (PKA) signaling cascade. Further, using functional magnetic resonance imaging (fMRI) and behavioral assessment, we show that d-serine is required for the potentiation of cognition by D3R blockade as revealed in a test of novel object recognition memory. Collectively, these results unveil a key role for d-serine in the dopaminergic neuromodulation of glutamatergic transmission and PFC activity, findings with clear relevance to the pathogenesis and treatment of diverse brain disorders involving alterations in dopamine-glutamate cross-talk.


Subject(s)
Dopamine/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/metabolism , Animals , Glutamic Acid/metabolism , Male , Mice , Mice, Knockout , Racemases and Epimerases/deficiency , Racemases and Epimerases/genetics , Receptors, Dopamine/metabolism , Schizophrenia , Synaptic Transmission/drug effects
4.
J Neurosci Res ; 98(8): 1561-1574, 2020 08.
Article in English | MEDLINE | ID: mdl-32468598

ABSTRACT

Dysfunctional social behavior is a major clinical feature of mood, anxiety, autism spectrum, and substance-related disorders, and may dramatically contribute to the poor outcome of these diseases. Nevertheless, the mechanisms underlying social behavior deficits are still largely unknown. The corticotropin-releasing factor (CRF) system, a major coordinator of the stress response, has been hypothesized to modulate social behavior. CRF signaling is mediated by two receptor types, termed CRF1 and CRF2 . Using the three-chamber task for sociability (i.e., preference for an unfamiliar conspecific vs. an object), this study demonstrates that CRF2 receptor null mutation (CRF2 -/-) reduces sociability in female mice but increases it in male mice. Both female and male CRF2 -/- mice display a preference for social odor cues over neutral cues, indicating that sex- and CRF2 receptor-dependent sociability is not due to altered olfaction or impaired social cues discrimination. Moreover, treatment with the CRF1 receptor-preferring antagonist, antalarmin, consistently induces sociability in non-social mice but disrupts it in social mice, independently of CRF2 receptor deficiency. Sex, CRF2 receptor deficiency, or antalarmin affect locomotor activity during the three-chamber test. However, throughout the study CRF1 and CRF2 receptor-linked sociability is independent of locomotor activity. The present findings highlight major functions for the CRF system in the regulation of social behavior. Moreover, they provide initial evidence of sex-linked roles for the CRF1 and the CRF2 receptor, emphasizing the importance of sex as a major biological variable to be taken into consideration in preclinical and clinical studies.


Subject(s)
Receptors, Corticotropin-Releasing Hormone/physiology , Sex Factors , Social Behavior , Animals , Anxiety , Female , Male , Mice , Mice, Inbred Strains , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Stress, Psychological
5.
Article in English | MEDLINE | ID: mdl-31669507

ABSTRACT

Poor sociability and aggressive behavior are major clinical features of opiate use disorders that may contribute to the establishment and maintenance of these harmful diseases. The present study investigated the long-term effects of chronic morphine administration and withdrawal upon social and aggressive behavior as well as the interrelationship between these two behaviors. Thus, social behavior was measured in C57BL/6J male mice 7, 21, 35 and 49 days after cessation of escalating morphine doses (20-100 mg/kg, i.p.) administered during 6 consecutive days, using the three-chamber task for sociability (i.e., preference for an unfamiliar conspecific vs. an object) and social novelty preference (i.e., preference for a novel vs. a familiar conspecific). Moreover, aggressive biting behavior towards an unfamiliar conspecific was assessed throughout the three-chamber tests. Opiate withdrawal increased both social approach and aggressive biting behavior. Moreover, in morphine-withdrawn, but not in control, mice social approach and aggressive behavior followed a similar time-course and positively correlated one with each other, suggesting that social interest was mainly driven by aggressiveness. Aggressive biting behavior was still elevated 49 days after the last morphine administration, revealing that opiate withdrawal is followed by long-lasting aggressiveness. Throughout, opiate withdrawal did not affect social novelty preference, ruling out a role for olfactory or social discrimination dysfunction in the elevated social approach and aggressive behavior. The present findings of very long-lasting aggressive behavior and aggression-driven social approach in opiate-withdrawn mice might help understanding the behavioral and brain underpinnings of poor sociability and aggressiveness commonly observed in opiate use disorders patients.


Subject(s)
Aggression/drug effects , Morphine/adverse effects , Social Behavior , Substance Withdrawal Syndrome/psychology , Animals , Male , Mice , Models, Animal , Time Factors
6.
Br J Pharmacol ; 175(9): 1504-1518, 2018 05.
Article in English | MEDLINE | ID: mdl-29406581

ABSTRACT

BACKGROUND AND PURPOSE: Poor social behaviour and vulnerability to stress are major clinical features of stimulant use disorders. The corticotropin-releasing factor (CRF) system mediates stress responses and might underlie substance use disorders; however, its involvement in social impairment induced by stimulant substances remains unknown. CRF signalling is mediated by two receptor types, CRF1 and CRF2 . In the present study we investigated the role of the CRF2 receptor in social behaviour deficits, vulnerability to stress and related brain alterations induced by cocaine administration and withdrawal. EXPERIMENTAL APPROACH: CRF2 receptor-deficient (CRF2 -/-) and littermate wild-type mice were repeatedly tested in the three-chamber task for sociability (i.e. preference for an unfamiliar conspecific vs. an object) and social novelty preference (SNP; i.e. preference for a novel vs. a familiar conspecific) before and after chronic cocaine administration. An in situ hybridization assay was used to assess gene expression of the stress-responsive arginine vasopressin (AVP) and oxytocin (OT) neuropeptides in the hypothalamus. KEY RESULTS: CRF2 receptor deficiency eliminated the sociability deficit induced by cocaine withdrawal. Moreover, CRF2 -/- mice did not show either the stress-induced sociability deficit or the increased AVP and OT expression associated with long-term cocaine withdrawal, indicating resilience to stress. Throughout, wild-type and CRF2 -/- mice displayed SNP, suggesting that cocaine withdrawal-induced sociability deficits were not due to impaired detection of social stimuli. CONCLUSIONS AND IMPLICATIONS: These findings demonstrate a central role for the CRF2 receptor in social behaviour deficits and biomarkers of vulnerability induced by cocaine withdrawal, suggesting new therapeutic strategies for stimulant use disorders.


Subject(s)
Cocaine/adverse effects , Receptors, Corticotropin-Releasing Hormone/deficiency , Social Behavior , Stress, Psychological/psychology , Substance Withdrawal Syndrome/psychology , Animals , Arginine Vasopressin/biosynthesis , Corticotropin-Releasing Hormone , Exploratory Behavior , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Oxytocin/biosynthesis , Receptors, Corticotropin-Releasing Hormone/genetics , Stress, Psychological/metabolism , Substance Withdrawal Syndrome/metabolism
7.
Neuropharmacology ; 117: 41-48, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28137450

ABSTRACT

Stimulant drugs produce reward but also activate stress-responsive systems. The corticotropin-releasing factor (CRF) and the related hypothalamus-pituitary-adrenal (HPA) axis stress-responsive systems are activated by stimulant drugs. However, their role in stimulant drug-induced reward remains poorly understood. Herein, we report that CRF1 receptor-deficient (CRF1-/-), but not wild-type, mice show conditioned place preference (CPP) responses to a relatively low cocaine dose (5 mg/kg, i.p.). Conversely, wild-type, but not CRF1-/-, mice display CPP responses to a relatively high cocaine dose (20 mg/kg, i.p.), indicating that CRF1 receptor-deficiency alters the rewarding effects of cocaine. Acute pharmacological antagonism of the CRF1 receptor by antalarmin also eliminates cocaine reward. Nevertheless, CRF1-/- mice display higher stereotypy responses to cocaine than wild-type mice. Despite the very low plasma corticosterone concentration, CRF1-/- mice show higher nuclear glucocorticoid receptor (GR) levels in the brain region of the hippocampus than wild-type mice. Full rescue of wild-type-like corticosterone and GR circadian rhythm and level in CRF1-/- mice by exogenous corticosterone does not affect CRF1 receptor-dependent cocaine reward but induces stereotypy responses to cocaine. These results indicate a critical role for the CRF1 receptor in cocaine reward, independently of the closely related HPA axis activity.


Subject(s)
Cocaine/pharmacology , Receptors, Corticotropin-Releasing Hormone/physiology , Animals , Circadian Rhythm , Cocaine/antagonists & inhibitors , Conditioning, Psychological/drug effects , Corticosterone/blood , Dose-Response Relationship, Drug , Hippocampus/metabolism , Male , Mice , Mice, Knockout , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Glucocorticoid/metabolism , Reward , Stereotyped Behavior/drug effects
8.
Neuropharmacology ; 105: 500-507, 2016 06.
Article in English | MEDLINE | ID: mdl-26907806

ABSTRACT

Opiate use disorders are associated with impaired cognitive function and altered stress-responsive systems. The corticotropin-releasing factor (CRF) system mediates stress responses via CRF1 and CRF2 receptors and may be implicated in substance use disorders. However, the specific role for each of the two known CRF receptor subtypes in cognitive impairment induced by opiate administration and withdrawal remains to be elucidated. In the present study, CRF1-/-, CRF2-/- and their respective wild-type mice are injected with escalating doses of morphine and cognitive function assessed by the novel object recognition (NOR) memory task throughout relatively long periods of opiate withdrawal. Early (2 days) phases of opiate withdrawal impair NOR memory in wild-type, CRF1-/- and CRF2-/- mice. However, the duration of opiate withdrawal-induced NOR memory deficits is prolonged in CRF1-/- but shortened in CRF2-/- mice, as compared to their respective wild-type mice, indicating opposite roles for the two CRF receptor subtypes. Nevertheless, following apparent recovery, exposure to an environmental stressor induces the reemergence of NOR memory deficits in long-term opiate-withdrawn wild-type but not CRF1-/- or CRF2-/- mice, indicating an essential role for both CRF receptor subtypes in stress vulnerability. These findings bring initial evidence of a complex physiopathological role for the CRF system in cognitive deficits and the long-lasting vulnerability induced by opiate drugs.


Subject(s)
Morphine/pharmacology , Opiate Alkaloids/pharmacology , Receptors, Corticotropin-Releasing Hormone/metabolism , Recognition, Psychology/drug effects , Substance Withdrawal Syndrome/metabolism , Animals , Cognition/drug effects , Cognition/physiology , Female , Memory Disorders/chemically induced , Memory Disorders/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Receptors, Corticotropin-Releasing Hormone/genetics , Recognition, Psychology/physiology , Resilience, Psychological , Stress, Psychological/metabolism , Substance Withdrawal Syndrome/psychology , Time Factors
9.
Neuropsychopharmacology ; 40(8): 1990-2000, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25672976

ABSTRACT

Vulnerability to stressful life events is a hallmark of drug dependence that may persist long after cessation of drug intake and dramatically fuel key clinical features, such as deregulated up-shifted motivational states and craving. However, to date, no effective therapy is available for reducing vulnerability to stressful events in former drug users and drug-dependent patients, mostly because of poor knowledge of the mechanisms underlying it. In this study, we report that genetic inactivation of the stress-responsive corticotropin-releasing factor receptor-2 (CRF2-/-) completely eliminates the reemergence of increased nonrewarded nose-pokes, reflecting up-shifted motivational states, triggered by ethological environmental stressors long after cessation of morphine administration in mice. Accordingly, CRF2 receptor deficiency completely abolishes the increase in biomarkers of synthesis of major brain motivational substrates, such as ventral tegmental area (VTA) dopamine (DA) and amygdala γ-aminobutyric acid (GABA) systems, associated with the stress-induced reemergence of up-shifted motivational states long after opiate withdrawal. Nevertheless, neither CRF2 receptor deficiency nor long-term opiate withdrawal affects amygdala CRF or hypothalamus CRF expression, indicating preserved brain stress-coping systems. Moreover, CRF2 receptor deficiency does not influence the locomotor or the anxiety-like effect of long-term opiate withdrawal. Thus, the present results reveal an essential and specific role for the CRF2 receptor in the stress-induced reemergence of up-shifted motivational states and related alterations in brain motivational systems long after opiate withdrawal. These findings suggest new strategies for the treatment of the severe and long-lasting vulnerability that inexorably follows drug withdrawal and hinder drug abstinence.


Subject(s)
Motivation/genetics , Opioid-Related Disorders/physiopathology , Receptors, Corticotropin-Releasing Hormone/deficiency , Substance Withdrawal Syndrome/etiology , Animals , Anxiety/genetics , Anxiety/physiopathology , Brain/metabolism , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Disease Models, Animal , Dopamine/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Reinforcement Schedule , Sex Factors , Substance Withdrawal Syndrome/pathology , Time Factors , gamma-Aminobutyric Acid/metabolism
10.
Int J Neuropsychopharmacol ; 17(12): 1969-79, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24800964

ABSTRACT

Psychostimulant drug abuse, dependence and withdrawal are associated with cognitive dysfunction and impact stress-sensitive systems. The corticotropin-releasing factor (CRF) system orchestrates stress responses via CRF1 and CRF2 receptors and is implicated in substance use disorders. However, CRF2 role in psychostimulant drug-induced cognitive dysfunction remains to be elucidated. In the present study, wild-type and CRF2-/- mice are injected with cocaine and memory assessed by the novel object recognition (NOR) task throughout relatively long periods of drug withdrawal. Following recovery from the drug-induced memory deficits, the mice are stressed prior to the NOR task and brain gene expression evaluated by in situ hybridization. Cocaine impairs NOR memory in wild-type and CRF2-/- mice. However, following cocaine withdrawal NOR memory deficits last less time in CRF2-/- than in wild-type mice. Furthermore, a relatively mild stressor induces the re-emergence of NOR deficits in long-term cocaine-withdrawn wild-type but not CRF2-/- mice. Cocaine-withdrawn mice show a genotype-independent higher c-fos expression in the NOR memory-relevant perirhinal cortex than drug-naïve mice. However neither genotype nor drug withdrawal affect the expression of tyrosine hydroxylase in the ventral tegmental area or the locus coeruleus and CRF in the central nucleus of the amygdala or the paraventricular nucleus of the hypothalamus, brain regions implicated in stress and drug responses. These data indicate a new role for the CRF2 receptor in cognitive deficits induced by cocaine withdrawal, both as regards to their duration and their re-induction by stress. Interestingly, prototypical brain stress systems other than CRF do not appear to be involved.


Subject(s)
Brain/physiopathology , Cocaine-Related Disorders/physiopathology , Memory Disorders/physiopathology , Receptors, Corticotropin-Releasing Hormone/deficiency , Stress, Psychological/physiopathology , Substance Withdrawal Syndrome/physiopathology , Animals , Brain/drug effects , Central Nervous System Stimulants/pharmacology , Cocaine/pharmacology , Gene Expression/drug effects , Male , Memory Disorders/chemically induced , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Recognition, Psychology/physiology , Resilience, Psychological , Stress, Psychological/etiology , Substance Withdrawal Syndrome/complications , Tyrosine 3-Monooxygenase/metabolism
11.
Psychopharmacology (Berl) ; 231(20): 3965-72, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24687410

ABSTRACT

RATIONALE: The intake of psychostimulant drugs may induce cognitive dysfunction and negative affective-like states, and is associated with increased activity of stress-responsive systems. The corticotropin-releasing factor (CRF) system mediates neuroendocrine, behavioural and autonomic responses to stressors, and might be implicated in substance-related disorders. CRF signalling is mediated by two receptor types, named CRF1 and CRF2. OBJECTIVES: The present study aims to elucidate the role for the CRF1 receptor in cognitive dysfunction and anxiety-like states induced by cocaine. RESULTS: The genetic inactivation of the CRF1 receptor (CRF1+/- and CRF1-/-) does not influence recognition memory in drug-naïve mice, as assessed by the novel object recognition (NOR) test. Moreover, the chronic administration of escalating doses of cocaine (5-20 mg/kg, i.p.) induces NOR deficits, which are unaffected by CRF1 receptor-deficiency. However, the same drug regimen reveals an anxiety-like vulnerability to cocaine in CRF1-/- but not in wild-type or CRF1+/- mice, as assessed by the elevated plus maze test. CONCLUSIONS: The present findings indicate dissociation of cognitive dysfunction and anxiety-like states induced by cocaine. Moreover, they unravel a novel mechanism of vulnerability to psychostimulant drugs.


Subject(s)
Anxiety/genetics , Central Nervous System Stimulants/pharmacology , Cocaine/pharmacology , Motor Activity/drug effects , Receptors, Corticotropin-Releasing Hormone/genetics , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Corticotropin-Releasing Hormone/pharmacology , Male , Mice , Mice, Knockout , Motor Activity/genetics , Recognition, Psychology/drug effects , Recognition, Psychology/physiology
12.
Neuropharmacology ; 73: 41-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23707482

ABSTRACT

Altered motivational processes are key features of drug dependence and withdrawal, yet their neural mechanisms remain largely unknown. The present study shows that genetic disruption of the corticotropin-releasing factor receptor-2 (CRF2-/-) does not impair motivation for palatable food in drug-naïve mice. However, CRF2 receptor-deficiency effectively reduces the increase in palatable food-driven motivation induced by opiate withdrawal. Indeed, both in male and female wild-type mice, withdrawal from escalating morphine doses (20-100 mg/kg) induces a dramatic and relatively long-lasting (6 days) increase in palatable food-driven operant behavior under a progressive ratio (PR) schedule of reinforcement. In contrast, either male or female morphine-withdrawn CRF2-/- mice show smaller and shorter (2 days) increases in motivation than wild-type mice. Nevertheless, CRF2 receptor-deficiency does not impair the ability to discriminate reinforced behavior prior to, during the partial opiate withdrawal periods occurring between morphine injections and following drug discontinuation, indicating preserved cognitive function. Moreover, CRF2 receptor-deficiency does not affect the ambulatory or body weight effects of intermittent morphine injections and withdrawal. These results provide initial evidence of a gender-independent and specific role for the CRF2 receptor in the motivational effects of opiate withdrawal.


Subject(s)
Morphine/pharmacology , Motivation/physiology , Receptors, Corticotropin-Releasing Hormone/physiology , Substance Withdrawal Syndrome/physiopathology , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Eating/drug effects , Eating/physiology , Female , Male , Mice , Motivation/drug effects , Mutation , Receptors, Corticotropin-Releasing Hormone/genetics , Reinforcement Schedule , Substance Withdrawal Syndrome/psychology
13.
Psychopharmacology (Berl) ; 221(4): 675-84, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22207240

ABSTRACT

RATIONALE: In drug-dependent individuals, the primary excessive motivation is for drugs. Studies also indicate altered interest for "natural" rewarding activities associated with motivational disorders that may be relevant to drug dependence. However, to date, the impact of drug dependence and withdrawal upon motivation for "natural" rewards remains unclear. METHODS AND OBJECTIVES: In the present study, we use a food-driven operant behavior paradigm to assess the impact of opiate intake and withdrawal upon the motivational properties of highly palatable food (HPF) in mice. RESULTS: Our findings indicate that early (8-h) opiate withdrawal does not affect either the motivational or the discriminative properties of HPF intake. However, starting 32 h after the last morphine injection, opiate withdrawal increases operant behavior aimed at obtaining HPF. The increased HPF-driven behavior lasts at least 12 days following opiate withdrawal, indicating long-lasting effects upon motivation. Using a paradigm of reward contingency reversal, we also address the impact of opiate withdrawal upon cognitive functions. Our results indicate that opiate withdrawal does not affect the ability to learn a new operant rule to obtain HPF. Indeed, opiate withdrawal ameliorates the acquisition of the new HPF-driven operant task, most probably due to the persistent and long-lasting increased motivation. Finally, analysis of ambulatory activity and body weight (BW) changes reveal that motivational and cognitive effects are totally independent of caloric and/or motor effects of opiate dosing and withdrawal. CONCLUSIONS: These results clearly demonstrate that excessive opiate intake and withdrawal produces dramatic and long-lasting motivational disorders relevant to drug dependence.


Subject(s)
Morphine Dependence/psychology , Morphine/administration & dosage , Reward , Substance Withdrawal Syndrome/psychology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Animals , Cognition/drug effects , Conditioning, Operant/drug effects , Eating/drug effects , Female , Mice , Morphine/adverse effects , Motivation/drug effects , Time Factors
14.
Int J Eat Disord ; 42(5): 402-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19130488

ABSTRACT

OBJECTIVE: Given the lack of reliable murine model of binge-like eating, we tried to induce this pathological behavior in mice. METHOD: We used an experimental protocol mimicking the etiological factors involved in the development of binge eating in humans, that is, food restriction, refeeding (R-R) in presence of high palatable food, and stress (S). RESULTS: Mice subjected to at least three cycles of R-R plus S (forced swimming stress), showed a binge-like behavior evident as early as 4 h, persisting 24 h after stress application and not associated to depressive-like behavior. However, after the third R-R/S cycle, food intakes of mice returned to normal levels. DISCUSSION: (i) at least three cycles of R-R plus S are required to promote abnormal eating in mice, (ii) this is not associated to depressive-like behaviors, and (iii) the enhanced pathological behavior showed a transient nature not persisting after the third R-R/S cycle.


Subject(s)
Bulimia , Disease Models, Animal , Animals , Energy Intake , Female , Food Deprivation/physiology , Food Preferences , Hunger , Hyperphagia , Mice , Mice, Inbred C57BL , Motivation , Random Allocation , Stress, Physiological , Stress, Psychological/physiopathology , Swimming , Taste
15.
Neuropsychopharmacology ; 33(12): 2878-87, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18288089

ABSTRACT

Escape from the extremely aversive opiate withdrawal symptoms powerfully motivates compulsive drug-seeking and drug-taking behaviors. The corticotropin-releasing factor (CRF) system is hypothesized to mediate the motivational properties of drug dependence. CRF signaling is transmitted by two receptor pathways, termed CRF(1) and CRF(2). To investigate the role for the CRF(2) receptor pathway in somatic opiate withdrawal, in the present study we used genetically engineered mice deficient in the CRF(2) receptor (CRF(2)-/-). We employed a novel, clinically relevant mouse model of 'spontaneous' opiate withdrawal as well as a classical opioid receptor antagonist (naloxone)-precipitated opiate withdrawal paradigm. To induce opiate dependence, mice were treated with intermittent escalating morphine doses (20-100 mg/kg, i.p.). We found that 8-128 h after the last opiate injection, CRF(2)-/- mice showed decreased levels of major somatic signs of spontaneous opiate withdrawal, such as paw tremor and wet dog shake, as compared to wild-type mice. Similarly, challenge with naloxone 2 h after the last morphine injection induced lower levels of paw tremor and wet dog shake in CRF(2)-/- mice as compared to wild-type mice. Despite the differences in somatic signs, wild-type and CRF(2)-/- mice displayed similar plasma corticosterone responses to opiate dosing and withdrawal, indicating a marginal role for the hypothalamus-pituitary-adrenal axis in the CRF(2) receptor mediation of opiate withdrawal. Our results unravel a novel role for the CRF(2) receptor pathway in opiate withdrawal. The CRF(2) receptor pathway might be a critical target of therapies aimed at alleviating opiate withdrawal symptoms and reducing relapse to drug intake.


Subject(s)
Brain Chemistry/drug effects , Brain Chemistry/genetics , Brain/drug effects , Opioid-Related Disorders/genetics , Receptors, Corticotropin-Releasing Hormone/genetics , Substance Withdrawal Syndrome/genetics , Animals , Brain/metabolism , Brain/physiopathology , Corticosterone/blood , Corticosterone/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Narcotic Antagonists/pharmacology , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/physiopathology , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/physiopathology , Time Factors
16.
Neuropharmacology ; 53(6): 733-40, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17884109

ABSTRACT

Improving rehabilitation after a severe tissue injury does not only require a reduction in pain, but also requires alleviation of negative affects, particularly anxiety. Although opioids remain unsurpassed analgesics to relieve moderate to severe pain, it has been shown that they also induce latent pain sensitization leading to long-lasting hyperalgesia via N-methyl-D-aspartate-(NMDA)-dependent pronociceptive systems. The present study evaluated the ability of nitrous oxide (N2O), a gas with NMDA antagonist properties, to prevent latent pain sensitization and long-term anxiety-like behavior (ALB) in rats with pain and opioid experiences. On D0, the pro-inflammatory drug carrageenan was injected in one hind paw of rats treated with fentanyl (4x100 microg/kg subcutaneously). Nociceptive threshold was evaluated with the paw pressure vocalization test. Rats were re-exposed to carrageenan or exposed to repeated non-nociceptive environmental stress (NNES) 2-3 weeks later. Rats were also challenged in the elevated plus-maze 2 weeks after fentanyl administration for evaluating ALB. The preventive effects of a single 4 h 50/50% N2O-O2 exposure performed on D0 was evaluated. Fifty percent N2O strongly reduced hyperalgesia induced by a first inflammation and its enhancement by fentanyl, and prevented exaggerated hyperalgesia induced by second inflammatory pain or NNES. Moreover, we provide first evidence that a high fentanyl dose induces long-term ALB 2 weeks after its administration. When associated with fentanyl, 50% N2O prevented such long-term ALB. These results suggest that a single exposure to N2O could improve post-injury pain management and facilitate rehabilitation especially when potent analgesics as opioids have to be used.


Subject(s)
Anxiety/prevention & control , Behavior, Animal/drug effects , Hyperalgesia/prevention & control , Nitrous Oxide/pharmacology , Pain/prevention & control , Analgesics, Opioid/pharmacology , Animals , Anxiety/chemically induced , Atmosphere Exposure Chambers , Disease Models, Animal , Fentanyl/pharmacology , Male , Pain/drug therapy , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Stress, Physiological/drug therapy
17.
Eur J Neurosci ; 25(11): 3398-405, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17553008

ABSTRACT

Altered motivational processes might participate to the physiopathology of eating-related disorders. The endogenous opioid system is thought to mediate the hedonic properties of food intake. To assess the role for the micro-opioid receptor (MOR) pathway in the motivational properties of food intake, in the present study we tested wild-type and MOR-deficient mice (MOR-/-) in a nose-poke operant paradigm for chow or sucrose pellets. To avoid confounding factors linked to food restriction/deprivation experience, mice were always provided with food ad libitum. Although less MOR-/- than wild-type mice initiated operant behaviour, under a fixed ratio-1 (FR-1) reinforcement schedule the two genotypes showed similar patterns of food-driven nose-poking, indicating preserved cognitive abilities in MOR-deficient mice. However, during FR-3 and progressive ratio (PR) reinforcement experiments, MOR-/- mice showed lower levels of nose-poking for either chow or sucrose pellets than wild-type mice, indicating a crucial role for the MOR pathway in the motivational properties of food intake. Moreover, under the PR reinforcement schedule mice nose-poking for sucrose pellets showed higher genotype-independent breakpoint levels than mice working for chow pellets, indicating that the MOR pathway is not essential for hedonic processing of palatable food intake. Finally, MOR-/- mice did not differ from wild-type mice in the rate of operant responding extinction, further supporting the notion of unaltered cognitive abilities in the MOR-deficient mice. The present findings strongly indicate that the MOR pathway mediates the motivational properties of food intake, but it is not essential for hedonic processing of ingestive behaviour.


Subject(s)
Eating/genetics , Motivation , Receptors, Opioid, mu/deficiency , Animals , Behavior, Animal , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Eating/drug effects , Extinction, Psychological , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Reinforcement Schedule , Reinforcement, Psychology , Sucrose/administration & dosage , Sweetening Agents/administration & dosage
18.
Neuron ; 53(4): 577-89, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17296558

ABSTRACT

Escape from the extremely stressful opiate withdrawal syndrome may motivate opiate seeking and taking. The corticotropin-releasing factor receptor-1 (CRF1) pathway mediates behavioral and endocrine responses to stress. Here, we report that genetic inactivation (CRF1-/-) as well as pharmacological antagonism of the CRF/CRF1 receptor pathway increased and prolonged the somatic expression of opiate withdrawal. Opiate-withdrawn CRF1-/- mice also showed aberrant CRF and dynorphin expression in the paraventricular nucleus of the hypothalamus (PVN) and the striatum, indicating profound impairments in stress-responsive brain circuitry. Intake of nonstressful amounts of corticosterone effectively reduced the exaggerated somatic reactions of CRF1-/- mice to opiate withdrawal. Exogenous corticosterone also restored "wild-type-like" patterns of CRF and dynorphin gene expression in the PVN and the striatum of opiate-withdrawn CRF1-/- mice, respectively. The present findings unravel a key role for the hypothalamus-pituitary-adrenal (HPA) system and brain extra-hypothalamic CRF/CRF1 receptor circuitry in somatic, molecular, and endocrine alterations induced by opiate withdrawal.


Subject(s)
Morphine/adverse effects , Receptors, Corticotropin-Releasing Hormone/physiology , Signal Transduction/physiology , Stress, Psychological/complications , Substance Withdrawal Syndrome/physiopathology , Analysis of Variance , Animals , Behavior, Animal/drug effects , Corticosterone/blood , Corticosterone/therapeutic use , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Disease Models, Animal , Dynorphins/genetics , Dynorphins/metabolism , Female , Mice , Mice, Knockout , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, Corticotropin-Releasing Hormone/deficiency , Signal Transduction/drug effects , Substance Withdrawal Syndrome/blood , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/genetics , Time Factors
19.
Behav Brain Res ; 170(1): 110-8, 2006 Jun 03.
Article in English | MEDLINE | ID: mdl-16580078

ABSTRACT

The opiate withdrawal syndrome powerfully motivates opiate seeking and abuse. Development of effective medications for opiate withdrawal symptoms is thus a primary research goal and heavily relies on improved experimental models. This study was carried out to establish a clinically relevant paradigm to assess somatic opiate withdrawal in mice. Female and male C57BL/6J mice were treated with saline or increasing morphine doses (10-50mg/kg or 20-100mg/kg) during 6 consecutive days and tested for the spontaneous expression of somatic opiate withdrawal signs 8, 32, 56 and 80 h after last drug administration. Contrary to opioid receptor antagonist-precipitated procedures, the spontaneous opiate withdrawal paradigm used here revealed interesting gender- and morphine dose-linked differences. In particular, 56 h after last morphine administration elevated global opiate withdrawal scores were still evident in female but not in male mice treated with 20-100mg/kg. The severity of somatic opiate withdrawal directly correlated with the prior cumulative morphine exposure. Timing of expression of somatic opiate withdrawal signs also varied as a function of both gender and morphine dose. For example, expression of paw tremors and wet dog shakes was earlier in opiate-withdrawn male than in female mice. Overall, these findings highlight the possibility to detect gender- and opiate dose-linked differences in the expression and duration of somatic opiate withdrawal using a clinically relevant research model. The behavioral paradigm described here may represent a more appropriate tool to investigate the neurobiological bases of opiate withdrawal as opposed to opioid receptor antagonist-precipitated opiate withdrawal procedures.


Subject(s)
Behavior, Animal/drug effects , Morphine Dependence/physiopathology , Morphine/toxicity , Substance Withdrawal Syndrome/physiopathology , Animals , Behavior, Animal/physiology , Brain/drug effects , Brain/physiopathology , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Motor Activity/drug effects , Motor Activity/physiology , Rats , Sex Factors
20.
Proc Natl Acad Sci U S A ; 102(51): 18649-54, 2005 Dec 20.
Article in English | MEDLINE | ID: mdl-16339307

ABSTRACT

The negative affective symptoms of opiate withdrawal powerfully motivate drug-seeking behavior and may trigger relapse to heroin abuse. To date, no medications exist that effectively relieve the negative affective symptoms of opiate withdrawal. The corticotropin-releasing factor (CRF) system has been hypothesized to mediate the motivational effects of drug dependence. The CRF signal is transmitted by two distinct receptors named CRF receptor-1 (CRF1) and CRF2. Here we report that genetic disruption of CRF1 receptor pathways in mice eliminates the negative affective states of opiate withdrawal. In particular, neither CRF1 receptor heterozygous (CRF1+/-) nor homozygous (CRF1-/-) null mutant mice avoided environmental cues repeatedly paired with the early phase of opiate withdrawal. These results were not due to altered associative learning processes because CRF1+/- and CRF1-/- mice displayed reliable, conditioned place aversions to environmental cues paired with the kappa-opioid receptor agonist U-50,488H. We also examined the impact of CRF1 receptor-deficiency upon opiate withdrawal-induced dynorphin activity in the nucleus accumbens, a brain molecular mechanism thought to underlie the negative affective states of drug withdrawal. Consistent with the behavioral indices, we found that, during the early phase of opiate withdrawal, neither CRF1+/- nor CRF1-/- showed increased dynorphin mRNA levels in the nucleus accumbens. This study reveals a cardinal role for CRF/CRF1 receptor pathways in the negative affective states of opiate withdrawal and suggests therapeutic strategies for the treatment of opiate addiction.


Subject(s)
Opioid-Related Disorders/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction , Substance Withdrawal Syndrome/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Dynorphins/genetics , Female , Gene Expression Regulation , Mice , Mice, Knockout , Nucleus Accumbens/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Corticotropin-Releasing Hormone/deficiency , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...