Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cerebellum ; 17(4): 428-437, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29450804

ABSTRACT

For many degenerative cerebellar diseases, currently, no effective treatment that would substantially restore cerebellar functions is available. Neurotransplantation could be a promising therapy for such cases. Nevertheless, there are still severe limitations for routine clinical use. The aim of the work was to assess volume and morphology and functional impact on motor skills of an embryonic cerebellar graft injected in the form of cell suspension in Lurcher mutant and wild-type mice of the B6CBA and C3H strains after a 6-month survival period. The grafts survived in the majority of the mice. In both B6CBA and C3H Lurcher mice, most of the grafts were strictly delimited with no tendency to invade the host cerebellum, while in wild-type mice, graft-derived Purkinje cells colonized the host's cerebellum. In C3H Lurcher mice, but not in B6CBA Lurchers, the grafts had smaller volume than in their wild-type counterparts. C3H wild-type mice had significantly larger grafts than B6CBA wild-type mice. No positive effect of the transplantation on performance in the rotarod test was observed. The findings suggest that the niche of the Lurcher mutant cerebellum has a negative impact on integration of grafted cells. This factor seems to be limiting for specific functional effects of the transplantation therapy in this mouse model of cerebellar degeneration.


Subject(s)
Brain Tissue Transplantation , Cerebellar Diseases/therapy , Cerebellum/embryology , Cerebellum/transplantation , Graft Survival , Neurodegenerative Diseases/therapy , Animals , Cerebellar Diseases/pathology , Cerebellar Diseases/physiopathology , Cerebellum/pathology , Disease Models, Animal , Female , Graft Survival/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Longitudinal Studies , Male , Mice, Inbred C3H , Mice, Inbred CBA , Mice, Neurologic Mutants , Mice, Transgenic , Motor Skills , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Rotarod Performance Test , Species Specificity
2.
Front Behav Neurosci ; 9: 32, 2015.
Article in English | MEDLINE | ID: mdl-25762910

ABSTRACT

In humans, optimism is a cognitive construct related to motivation; optimists exert effort, whereas pessimists disengage from effort. In this study, using a recently developed ambiguous-cue interpretation (ACI) paradigm we took the unique opportunity to investigate whether "optimism" as a trait is correlated with motivation in rodents. In a series of ACI tests (cognitive bias screening, CBS), we identified rats displaying "pessimistic" and "optimistic" traits. Subsequently, we investigated the trait differences in the motivation of these rats to gain reward and to avoid punishment using a progressive ratio (PR) schedule of reinforcement paradigm. Although "optimistic" and "pessimistic" animals did not differ in their motivation to avoid punishment, the "optimistic" rats were significantly more motivated to gain reward than their "pessimistic" conspecifics. For the first time, we showed an association between cognitive judgment bias and motivation in an animal model. Because both investigated processes are closely related to mental health and wellbeing, our results may be valuable for preclinical modeling of many psychiatric disorders.

3.
Psychopharmacology (Berl) ; 232(3): 651-60, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25116482

ABSTRACT

RATIONALE: Animal models of mania lack genuine cognitive parameters. The present gold standard of mania models, amphetamine-induced hyperlocomotion, is rather unspecific and does not necessarily target its cardinal symptoms. Therefore, alternative behavioral markers that are sensitive to stimulants are required. OBJECTIVES: In the present study, by combining the psychostimulant-induced model of mania in rodents with the recently developed ambiguous-cue interpretation (ACI) tests, we investigated the effects of chronic administration of D-amphetamine and cocaine on the cognitive judgment bias of rats. METHODS: To accomplish this goal, in two separate experiments, previously trained animals received chronic, daily injections of either D-amphetamine (2 mg/kg) or cocaine (10 mg/kg) for 2 weeks and were subsequently tested with the ACI procedure. RESULTS: Chronic treatment with both psychostimulants did not make rats more "optimistic." CONCLUSIONS: The results are discussed in terms of behavioral and pharmacological actions of the tested compounds and their implications for modeling mania in animals.


Subject(s)
Behavior, Animal/drug effects , Bipolar Disorder/chemically induced , Bipolar Disorder/physiopathology , Cocaine/pharmacology , Dextroamphetamine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Judgment/drug effects , Animals , Cocaine/administration & dosage , Dextroamphetamine/administration & dosage , Disease Models, Animal , Dopamine Uptake Inhibitors/administration & dosage , Male , Rats , Rats, Sprague-Dawley
4.
Pharmacol Rep ; 64(5): 1091-104, 2012.
Article in English | MEDLINE | ID: mdl-23238466

ABSTRACT

BACKGROUND: Immediate early gene (IEG) induction elicited by drugs of abuse may contribute to development of plastic changes in the brain responsible for drug-induced behavioral changes leading to addiction. The aim of the present study was to characterize the changes in IEG expression in the striatum and nucleus accumbens produced by an acute or chronic administration of morphine. METHODS: In order to search for a possible relationship between morphine-induced IEG expression and behavior, the experiment was performed on two inbred strains of mice, C57BL/6J and DBA/2J, which differ markedly in their sensitivity to the rewarding and locomotor stimulatory actions of opiates. Gene expression was assessed using RT-PCR and DNA microarrays. RESULTS: The experiments demonstrated a prolonged or a delayed up-regulation of 14 IEG in the striatum at 4 h after morphine administration. Among them, a cluster of 8 genes, including 6 inducible transcription factors (c-fos, fra-2, junB, zif268 (egr1), egr2, NGFI-B) and 2 effector IEG (arc and mkp1) seemed to be regulated in concert in response to morphine. This group of genes was induced to a greater degree after chronic than acute morphine administration selectively in C57BL/6J mice and the difference bore apparently no relationship to opiate-produced locomotor activation. The strain-selective regulation was also demonstrated for cyclin L2 and tPA after an acute morphine injection. CONCLUSIONS: Our data indicate that morphine up-regulates many IEG in the mouse striatum at a strikingly delayed time-point and that these changes are genotype-dependent. They also suggest inter-strain differences in the development of striatal neuroadaptations to chronic morphine treatment.


Subject(s)
Corpus Striatum/drug effects , Genes, Immediate-Early , Morphine/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Corpus Striatum/metabolism , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Minocycline/pharmacology , Motor Activity/drug effects , Pyrroles/pharmacology , Species Specificity
5.
Int J Neuropsychopharmacol ; 15(10): 1457-71, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22040728

ABSTRACT

The brain's noradrenergic system is involved in the development of behaviours induced by drugs of abuse, e.g. dependence and withdrawal, and also reward or psychomotor effects. To investigate how noradrenergic system activity is controlled in the context associated with drug-induced behaviours, we generated a Cre/loxP mouse model in which the essential glutamate NMDA receptor subunit NR1 is ablated in cells expressing dopamine ß-hydroxylase (Dbh). As a result, the noradrenergic cells in NR1DbhCre mice lack the NMDA receptor-dependent component of excitatory post-synaptic currents. The mutant mice displayed no obvious behavioural alterations, had unchanged noradrenaline content and mild increase in dopamine levels in the nucleus accumbens. Interestingly, NR1DbhCre animals did not develop morphine-induced psychomotor sensitization. However, when the morphine injections were preceded by treatment with RX821002, an antagonist of α2-adrenergic receptors, the development of sensitization was restored. Conversely, pretreatment with clonidine, an agonist of α2-adrenergic receptors, blocked development of sensitization in wild-type mice. We also found that while the development of tolerance to morphine was normal in mutant mice, withdrawal symptoms were attenuated. These data reveal that NMDA receptors on noradrenergic neurons regulate development of opiate dependence and psychomotor sensitization, by controlling drug-induced noradrenaline signalling.


Subject(s)
Adrenergic Neurons/metabolism , Carrier Proteins/biosynthesis , Glutamic Acid/physiology , Morphine Dependence/metabolism , Nerve Tissue Proteins/biosynthesis , Psychomotor Performance/physiology , Adrenergic Neurons/drug effects , Adrenergic alpha-Antagonists/pharmacology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morphine Dependence/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Organ Culture Techniques , Psychomotor Performance/drug effects , Receptors, N-Methyl-D-Aspartate
6.
Psychopharmacology (Berl) ; 208(2): 291-300, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19997907

ABSTRACT

RATIONALE: Vulnerability to drug abuse disorders is determined not only by environmental but also by genetic factors. A body of evidence suggests that endogenous opioid peptide systems may influence rewarding effects of addictive substances, and thus, their individual expression levels may contribute to drug abuse liability. OBJECTIVES: The aim of our study was to assess whether basal genotype-dependent brain expression of opioid propeptides genes can influence sensitivity to morphine reward. METHODS: Experiments were performed on inbred mouse strains C57BL/6J, DBA/2J, and SWR/J, which differ markedly in responses to morphine administration: DBA/2J and SWR/J show low and C57BL/6J high sensitivity to opioid reward. Proenkephalin (PENK) and prodynorphin (PDYN) gene expression was measured by in situ hybridization in brain regions implicated in addiction. The influence of the kappa opioid receptor antagonist nor-binaltorphimine (nor-BNI), which attenuates effects of endogenous PDYN-derived peptides, on rewarding actions of morphine was studied using the conditioned place preference (CPP) paradigm. RESULTS: DBA/2J and SWR/J mice showed higher levels of PDYN and lower levels of PENK messenger RNA in the nucleus accumbens than the C57BL/6J strain. Pretreatment with nor-BNI enhanced morphine-induced CPP in the opioid-insensitive DBA/2J and SWR/J strains. CONCLUSIONS: Our results demonstrate that inter-strain differences in PENK and PDYN genes expression in the nucleus accumbens parallel sensitivity of the selected mouse strains to rewarding effects of morphine. They suggest that high expression of PDYN may protect against drug abuse by limiting drug-produced reward, which may be due to dynorphin-mediated modulation of dopamine release in the nucleus accumbens.


Subject(s)
Behavior, Addictive/genetics , Behavior, Animal , Enkephalins/genetics , Morphine Dependence/genetics , Nucleus Accumbens/metabolism , Protein Precursors/genetics , Animals , Behavior, Animal/drug effects , Conditioning, Psychological , Cues , Enkephalins/metabolism , Gene Expression Regulation , Genotype , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Morphine Dependence/metabolism , Morphine Dependence/prevention & control , Morphine Dependence/psychology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Nucleus Accumbens/drug effects , Phenotype , Protein Precursors/metabolism , RNA, Messenger/metabolism , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/metabolism , Reward , Species Specificity
7.
Psychopharmacology (Berl) ; 207(2): 245-55, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19787337

ABSTRACT

RATIONALE: Interstrain differences in the motivational properties of morphine and heroin have been previously reported in mice, suggesting the involvement of a genotype-dependent modulation of the rewarding effects of opiates. Yet, interstrain differences in the motivational effects of naloxone have not been described. OBJECTIVES: The aim of our study was to examine genotype modulation of the motivational effects of opiates in inbred stains of mice with known, distinct, opiate-induced phenotypes, and morphine-induced striatal transcriptional responses. METHODS: We studied the rewarding properties of morphine (5, 10, and 20 mg/kg i.p.) and heroin (1, 5, and 10 mg/kg i.p.) in conditioned place preference (CPP) as well as the aversive properties of naloxone (1, 10, and 20 mg/kg i.p.) in the conditioned place aversion (CPA) paradigm in C57Bl/6J (C57), DBA/2J (DBA), and SWR/J (SWR) inbred strains of mice. RESULTS: Our results show that morphine and heroin as well as naloxone induce CPP and CPA, respectively, in a genotype- and dose-dependent manner in these studied inbred strains of mice. Interestingly, C57 mice are the most sensitive in the case of the rewarding properties of morphine and heroin but are the least sensitive to the aversive effects of naloxone, whereas the DBA strain exhibit the opposite behavioral effects. CONCLUSIONS: We suggest that motivational homeostasis can be modulated by mu opioid receptors in mice, with the C57 mice representing a genotype that is more sensitive to processes related to rewards, whereas the genotype of DBA is more sensitive to aversion.


Subject(s)
Heroin/pharmacology , Morphine/pharmacology , Naloxone/pharmacology , Reward , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Conditioning, Psychological/drug effects , Dose-Response Relationship, Drug , Genotype , Heroin/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Morphine/administration & dosage , Motivation/drug effects , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Species Specificity
8.
Pharmacol Biochem Behav ; 90(4): 512-6, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18474394

ABSTRACT

The proteins of Fos family are a potential candidate to link molecular mechanisms of morphine action with behavioural effects such as morphine-induced reward, dependence and tolerance. We used both male and female mice lacking fosB gene to study its contribution to morphine effects. Morphine analgesia (tail-flick test) and hypothermia were studied using morphine at cumulative doses in morphine-naive and morphine-tolerant (tolerance induced by 24 h prior 100 mg/kg morphine administration) mice. FosB -/- mice, as compared to fosB +/+ mice, developed enhanced tolerance to morphine-induced analgesia. No effects of genotype or gender on tolerance to morphine-induced hypothermia were observed. These results suggest that fosB may be involved in the development of tolerance to morphine analgesia but not hypothermia. The gender study implicates that lack of FosB proteins in female fosB -/- mice enhanced morphine analgesic potency. In conclusion, we show that fosB gene is important to analgesia but not hypothermia phenotype indicating its role in morphine effects.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Proto-Oncogene Proteins c-fos/physiology , Animals , Body Temperature/drug effects , Dose-Response Relationship, Drug , Drug Tolerance , Female , Hypothermia/chemically induced , Hypothermia/genetics , Male , Mice , Mice, Knockout , Pain Measurement/drug effects , Pain Threshold/drug effects , Proto-Oncogene Proteins c-fos/genetics , Sex Characteristics
9.
Behav Brain Res ; 190(2): 212-7, 2008 Jul 19.
Article in English | MEDLINE | ID: mdl-18407360

ABSTRACT

The immediate early genes (IEGs) have been suggested to be implicated in mechanisms of addiction, as well as in learning and memory processes. fosB, which belongs to IEG, has been reported to have pleiotropic impact on response to psychoactive drugs, as well as motivational and stress-related behaviours. In the present study, we used mice with constitutive knock-out of fosB in order to study fosB role in mouse phenotype. We studied rewarding properties of morphine (10mg/kg i.p.) in conditioned place preference (CPP) paradigm. Additionally, we studied fosB role in spatial memory and spatial working memory using elevated plus maze model of spatial learning (EPMSL) and delayed non-match to place task (DNMTP). In further studies, locomotor, depressive-like and anxiety-like behaviours were measured. Rewarding effects of morphine in fosB -/- mice were abolished whereas spatial learning was impaired. On the other hand, we found no significant differences in locomotor activity, depression-like and anxiety-like behaviours. In summary, our results indicate that mice lacking fosB are less sensitive to rewarding properties of morphine and display spatial memory impairment and suggest involvement of fosB and its proteins in motivational aspects of reinforcers as well as in learning and memory processes.


Subject(s)
Association Learning/drug effects , Maze Learning/drug effects , Memory, Short-Term/drug effects , Morphine/pharmacology , Narcotics/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Analysis of Variance , Animals , Association Learning/physiology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Male , Maze Learning/physiology , Memory, Short-Term/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Motivation , Motor Activity/drug effects , Motor Activity/physiology , Proto-Oncogene Proteins c-fos/genetics , Reinforcement, Psychology , Spatial Behavior/drug effects , Spatial Behavior/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...