Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 238: 119-23, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23085477

ABSTRACT

Brain regional analyses of total GluA1 and GluA1-pSer(845) were used to delineate plasticity of the AMPA receptor in conjunction with cocaine-cue extinction learning. Rats were trained to self-administer cocaine paired with a 2-s light cue and later underwent a single 2 h extinction session for which cocaine was withheld but response-contingent cues were presented. Control groups received yoked-saline sessions or received cocaine self-administration training without undergoing extinction training. Extinction-related increases and decreases, respectively, in total GluA1 were observed in the ventromedial prefrontal cortex (vmPFC) and basolateral amygdala (BLA). Phosphorylation of GluA1 at Ser(845) was increased in the vmPFC and nucleus accumbens (NAc). Though total GluA1 did not change in NAc, there was a positive association between the number of responses during extinction training and the magnitude of total GluA1 in NAc. No significant changes were evident in the dorsal hippocampus. We conclude that the BLA and vmPFC, in particular, appear to be loci for the inhibition of learned behavior induced via extinction training, but each site may have different signaling functions for cocaine-cue extinction learning.


Subject(s)
Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Extinction, Psychological/drug effects , Receptors, AMPA/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Behavior, Addictive/metabolism , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Cues , Extinction, Psychological/physiology , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Phosphorylation , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Self Administration
2.
Behav Brain Res ; 234(1): 100-6, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22721675

ABSTRACT

Extinguishing abnormally strengthened learned responses to cues associated with drugs of abuse remains a key tactic for alleviating addiction. To assist in developing pharmacotherapies to augment exposure therapy for relapse prevention, investigation into neurobiological underpinnings of drug-cue extinction learning is needed. We used regional analyses of c-Fos and GluR2 protein expression to delineate neural activity and plasticity that may be associated with cocaine-cue extinction learning. Rats were trained to self-administer cocaine paired with a light cue, and later underwent a single 2h extinction session for which cocaine was withheld but response-contingent cues were presented (cocaine-cue extinction). Control groups consisted of rats yoked to animals self-administering cocaine and receiving saline non-contingently followed by an extinction session, or rats trained to self-administer cocaine followed by a no-extinction session for which levers were retracted, and cocaine and cues were withheld. Among 11 brain sites examined, extinction training increased c-Fos expression in basolateral amygdala and prelimbic prefrontal cortex of cocaine-cue extinguished rats relative to both control conditions. In dorsal subiculum and infralimbic prefrontal cortex, extinction training increased c-Fos expression in both cocaine-cue and saline-cue extinguished rats relative to the no-extinction control condition. GluR2 protein expression was not altered in any site examined after extinction or control training. Findings suggest that basolateral amygdala and prelimbic prefrontal cortex neurons are activated during acquisition of cocaine-cue extinction learning, a process that is independent of changes in GluR2 abundance. Other sites are implicated in processing the significance of cues that are present early in extinction training.


Subject(s)
Brain/drug effects , Cocaine/pharmacology , Extinction, Psychological/drug effects , Learning/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Animals , Behavior, Addictive , Behavior, Animal/drug effects , Brain/metabolism , Male , Rats , Rats, Wistar , Receptors, AMPA/metabolism
3.
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
4.
Alcohol ; 19(2): 157-62, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10548160

ABSTRACT

The role of adenosine receptor-mediated signaling was examined in the alcohol withdrawal syndrome. CD-1 mice received a liquid diet containing ethanol (6.7%, v/v) or a control liquid diet that were abruptly discontinued after 14 days of treatment. Mice consuming ethanol showed a progressive increase in signs of intoxication throughout the drinking period. Following abrupt discontinuation of ethanol diet, mice demonstrated reversible signs of handling-induced hyperexcitability that were maximal between 5-8 h. Withdrawing mice received treatment with adenosine receptor agonists at the onset of peak withdrawal (5.5 h) and withdrawal signs were blindly rated (during withdrawal hours 6 and 7). Adenosine A1-receptor agonist R-N6(phenylisopropyl)adenosine (0.15 and 0.3 mg/ kg) reduced withdrawal signs 0.5 and 1.5 h after drug administration in a dose-dependent fashion. Adenosine A2A-selective agonist 2-p-(2-carboxyethyl)phenylethyl-amino-5'-N-ethylcarboxamidoadenosine (0.3 mg/kg) reduced withdrawal signs at both time points. In ethanol-withdrawing mice, there were significant decreases in adenosine transporter sites in striatum without changes in cortex or cerebellum. In ethanol-withdrawing mice, there were no changes in adenosine A1 and A2A receptor concentrations in cortex, striatum, or cerebellum. There appears to be a role for adenosine A1 and A2A receptors in the treatment of the ethanol withdrawal syndrome. Published by Elsevier Science Inc.


Subject(s)
Adenosine/analogs & derivatives , Central Nervous System Depressants/adverse effects , Ethanol/adverse effects , Phenethylamines/therapeutic use , Purinergic P1 Receptor Agonists , Receptors, Purinergic P1/drug effects , Substance Withdrawal Syndrome/drug therapy , Adenosine/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Central Nervous System Depressants/administration & dosage , Ethanol/administration & dosage , Male , Mice , Receptor, Adenosine A2A , Receptors, Purinergic P1/physiology , Substance Withdrawal Syndrome/psychology , Thioinosine/analogs & derivatives , Thioinosine/metabolism
5.
Neurosci Lett ; 273(3): 147-50, 1999 Oct 08.
Article in English | MEDLINE | ID: mdl-10515180

ABSTRACT

We examined the effects of chronic in vivo antipsychotic drug treatments on G protein function and regulation. Mice were treated with typical antipsychotic haloperidol (6 mg/kg per day) and atypical agent olanzapine (20 mg/kg per day) for 14 days via mini-osmotic pumps. G protein-activated adenylyl cyclase activity in brain tissues was measured in the presence of guanine nucleotide analogue guanosine-5'-O(3-thiotriphosphate) tetralithium salt, or GTPgammaS. In frontal cortex, haloperidol treatment produced 21% increases in the GTPgammaS -mediated adenylyl cyclase Emax value (vs. vehicle controls) while olanzapine produced 20% reductions in this value (vs. controls); these effects were significant. In striatum, olanzapine treatment produced significant 31 and 27% decreases in Emax values compared with vehicle and haloperidol treatment, respectively. Chronic haloperidol treatment produced significant 24% reductions in the immunoreactivity of cortical, but not striatal, Gialpha1,2 subunits. There were no effects of chronic olanzapine treatment on G(i)alpha1,2 levels and no effects of either antipsychotic on G(s)alpha, levels. Chronic haloperidol and olanzapine treatments differentially regulate G protein-mediated adenylyl cyclase responses in brain regions possibly relating to their unique effects on G protein-coupled receptors.


Subject(s)
Adenylyl Cyclases/drug effects , Antipsychotic Agents/pharmacology , GTP-Binding Proteins/drug effects , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Haloperidol/pharmacology , Pirenzepine/analogs & derivatives , Adenylyl Cyclases/metabolism , Animals , Benzodiazepines , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , GTP-Binding Proteins/metabolism , Male , Mice , Olanzapine , Pirenzepine/pharmacology
6.
Brain Res ; 804(1): 52-62, 1998 Aug 31.
Article in English | MEDLINE | ID: mdl-9729274

ABSTRACT

Regulation of post-receptor signaling provides a mechanism of adaptation to chronic psychotropic drug treatment. In this study, the regulation of guanine nucleotide binding proteins (G proteins) and G protein-stimulated adenylyl cyclase activity was examined in brain regions of caffeine-tolerant and -dependent mice. Chronic caffeine doses were administered via mini-osmotic pumps over 7 days at 0, 42, 85 and 125 mg kg-1 day-1. These chronic caffeine doses were linearly correlated with plasma caffeine concentrations. In behavioral studies, the stimulant effects of acute caffeine on motor activity were significantly diminished in a dose-dependent manner after chronic caffeine, suggesting the development of tolerance. Abrupt discontinuation of chronic caffeine treatment (at 85 and 125 mg kg-1 day-1) produced a dose-dependent and reversible reduction in motor activity 24 h later, suggestive of a caffeine withdrawal syndrome. Utilizing quantitative immunoblotting methods, we found that hippocampal Gialpha1,2 and Gialpha3 subunits were significantly reduced by 20.2% and 11.1%, respectively, in caffeine tolerant/dependent mice (caffeine 125 mg kg-1 day-1 vs. vehicle controls). Decreases in inhibitory G protein subunit concentrations in hippocampus were accompanied by a significant increase (by 21%) in hippocampal G protein function, as measured by guanine nucleotide-stimulated adenylyl cyclase activity, in caffeine-treated mice. This same caffeine treatment also produced significant decreases in cortical Gsalpha subunits of 14.0%. Since short-term caffeine treatment has been shown to reduce adenylyl cyclase activity, chronic caffeine treatment could produce adaptive increases in G protein-stimulated adenylyl cyclase to oppose this effect via G protein regulation.


Subject(s)
Adenylyl Cyclases/metabolism , Brain/metabolism , Caffeine/pharmacology , GTP-Binding Proteins/metabolism , Substance-Related Disorders/metabolism , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Drug Tolerance , Guanylyl Imidodiphosphate/metabolism , Male , Mice , Mice, Inbred Strains , Motor Activity/drug effects , Osmolar Concentration , Substance Withdrawal Syndrome/physiopathology , Tissue Distribution
7.
Brain Res ; 788(1-2): 104-10, 1998 Mar 30.
Article in English | MEDLINE | ID: mdl-9554971

ABSTRACT

Previous research has demonstrated that acute and chronic opiate treatment alters receptor- and postreceptor-mediated adenylyl cyclase activity. This study examined the regulation of G protein- and forskolin-mediated adenylyl cyclase activity in mouse striatum and cortex after short- and long-term opiate exposure. To directly measure adenylyl cyclase enzymatic activity, assays were done in the presence of catalytic site activator forskolin. To measure G protein-mediated adenylyl cyclase activity, assays were performed in the presence of non-hydrolyzable guanosine 5'-triphosphate (GTP) analogue, 5'-guanylyl-imidodiphosphate. Short-term in vitro morphine exposure produced reductions in forskolin-stimulated adenylyl cyclase activity in striatal and cortical tissues. Long-term morphine treatment in mice was performed via morphine- or placebo-pellet implantation for 72 h; this treatment has been shown to produce opiate dependence and withdrawal. In both opiate-dependent and opiate withdrawing mice (1 h post-naloxone induction), there were significant increases in G protein-mediated adenylyl cyclase activity in the striatum (vs. controls). In opiate-dependent mice, there was an decrease in G protein-mediated adenylyl cyclase activity in cortex. In opiate-dependent mice, there were no changes in forskolin-stimulated adenylyl cyclase in the striatum or cortex. Increases in striatal G protein-mediated adenylyl cyclase could represent a compensatory adaptation that opposes the persistent inhibition of adenylyl cyclase by chronic opiate treatment contributing to the expression of opiate dependence and withdrawal.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Proteins/physiology , Morphine Dependence/enzymology , Morphine/adverse effects , Substance Withdrawal Syndrome , Telencephalon/drug effects , Analysis of Variance , Animals , Cerebral Cortex/drug effects , Colforsin/pharmacology , Corpus Striatum/drug effects , Logistic Models , Male , Mice , Mice, Inbred Strains , Telencephalon/enzymology
8.
Brain Res ; 763(2): 215-20, 1997 Jul 25.
Article in English | MEDLINE | ID: mdl-9296562

ABSTRACT

Opioid-adenosine interactions have been demonstrated at both cellular and behavioral levels. Short-term morphine treatment has been shown to enhance adenosine release in brain and spinal tissues. Since adenosine uptake and release is regulated by a nitrobenzylthioinosine-sensitive adenosine transporter, we examined the effects of morphine treatment on this transporter-binding site. Adenosine transporter-binding sites were examined using equilibrium binding studies with [3H]nitrobenzylthioinosine in brain regions of morphine-treated mice. A 72-hour morphine pellet implantation procedure, which previously produced up-regulation of central adenosine A1 receptors and created a state of opiate dependence [G.B. Kaplan, K.A. Leite-Morris and M.T. Sears, Alterations in adenosine A receptors in morphine dependence, Brain Res., 657 (1994) 347-350], was used in this current study. This chronic morphine treatment significantly increased adenosine transporter-binding site concentrations in striatum and hypothalamus by 12 and 37%, respectively, compared to vehicle pellet implantation. No effects of morphine treatment were demonstrated in cortex, hippocampus, brainstem or cerebellum. In behavioral studies, mice receiving this same chronic morphine or vehicle treatment were given saline or morphine injections (40 or 50 mg/kg i.p.) followed by ambulatory activity monitoring. In the chronic vehicle treatment group, morphine injections significantly stimulated ambulatory activity while in the chronic morphine treatment group there was no such stimulation by acute morphine, suggestive of opiate tolerance. Morphine-induced up-regulation of striatal and hypothalamic adenosine transporter sites could potentially alter extracellular adenosine release and adenosine receptor activation and mediate aspects of opiate tolerance and dependence.


Subject(s)
Adenosine/metabolism , Carrier Proteins/metabolism , Corpus Striatum/chemistry , Hypothalamus/chemistry , Membrane Proteins/metabolism , Morphine/pharmacology , Narcotics/pharmacology , Animals , Binding Sites/drug effects , Binding Sites/physiology , Brain Stem/chemistry , Brain Stem/metabolism , Cerebellum/chemistry , Cerebellum/metabolism , Corpus Striatum/metabolism , Drug Tolerance , Hypothalamus/metabolism , Male , Mice , Mice, Inbred Strains , Nucleoside Transport Proteins , Up-Regulation/drug effects
9.
Brain Res ; 657(1-2): 347-50, 1994 Sep 19.
Article in English | MEDLINE | ID: mdl-7820640

ABSTRACT

The possibility that central adenosine A1 and A2a receptors mediate opiate dependence was examined in morphine-treated mice using radioligand binding methods. Mice treated with morphine for 72 h demonstrated significant increases in naloxone precipitated abstinence behaviors of jumping, wet-dog shakes, teeth chattering, forepaw trends, forepaw tremors and diarrhea compared to vehicle-treated mice. Increased concentrations of cortical adenosine A1 receptor sites, but not striatal adenosine A2a sites, were found in saturation binding studies from morphine-dependent mice. Decreases in cortical A1 agonist binding affinity values along with increases in agonist binding sites were demonstrated in competition binding studies. These results suggest that adaptive changes of upregulation and sensitization of adenosine A1 receptors play a role in mediating the opiate abstinence syndrome.


Subject(s)
Morphine Dependence/physiopathology , Receptors, Purinergic P1/physiology , Animals , Binding, Competitive , Male , Mice , Morphine Dependence/metabolism , Morphine Dependence/psychology , Radioligand Assay , Receptors, Purinergic P1/metabolism , Substance Withdrawal Syndrome/physiopathology
10.
J Immunol ; 147(8): 2717-27, 1991 Oct 15.
Article in English | MEDLINE | ID: mdl-1918986

ABSTRACT

The role of transforming growth factor beta 1 (TGF-beta 1) in regulating NK and T cell proliferation during acute viral infections was investigated. After infection of mice with lymphocytic choriomeningitis virus, NK cell proliferation peaks on day 3 and subsides by days 5 to 7 postinfection, whereas T cell proliferation peaks on day 7 and declines by days 9 to 14 postinfection. As TGF-beta 1 has been shown to inhibit lymphocyte proliferation in culture, the production and function of TGF-beta 1 during infection was evaluated in this model. Northern blot analysis demonstrated that the accumulation of TGF-beta 1 transcripts remained relatively constant in total splenic leukocytes during infection. The Mv 1 Lu mink lung cell bioassay was modified and used to evaluate the production of biologically active TGF-beta 1 during infection. Media conditioned with splenic leukocytes isolated from infected mice contained factors that inhibited DNA synthesis by the Mv 1 Lu cells. Low levels of inhibition were observed with conditioned media prepared on day 3 postinfection and high levels of inhibition were observed with conditioned media prepared on days 5 through 14 postinfection. Neutralization with antibodies specific for TGF-beta 1 demonstrated that TGF-beta 1 contributed to the inhibitory activity. As TGF-beta 1 was produced at times coinciding with the decline in NK cell proliferation, the TGF-beta 1 sensitivity of in vivo-elicited NK cells was evaluated. In vitro and in vivo studies demonstrated that NK cell proliferation was extremely sensitive to inhibition by TGF-beta 1. In culture, TGF-beta 1 had an ID50 of 8 pg/ml for inhibiting DNA synthesis by blast NK cells. In vivo, administration of a total of 0.18 micrograms of TGF-beta 1 resulted in a 93% inhibition of NK cell-mediated lytic units per spleen on day 3 postinfection. The inhibition was a result of a block in NK cell proliferation, as administration of TGF-beta 1 profoundly suppressed the appearance of blast size NK cells and the incorporation of [3H] thymidine by NK cell-enriched, blast lymphocyte populations on day 3 postinfection. In contrast to NK cell proliferation, T cell proliferation was not inhibited by up to 100-fold higher concentrations of the factor in vitro or in vivo. Taken together, these data demonstrate that TGF-beta 1 is an important regulator of NK cell proliferation in vivo. Furthermore, the results indicate that differential sensitivity to TGF-beta 1 may contribute to the coordination of NK and T cell responses during viral infections.


Subject(s)
Killer Cells, Natural/immunology , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/physiology , Acute Disease , Animals , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , RNA, Messenger/analysis , T-Lymphocytes, Cytotoxic/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology
11.
J Immunol ; 146(6): 1955-63, 1991 Mar 15.
Article in English | MEDLINE | ID: mdl-1672337

ABSTRACT

The murine immune response to lymphocytic choriomeningitis virus (LCMV) infection involves the activation of CD8+, class I MHC-restricted and virus-specific CTL. At times coinciding with CTL activation, high levels of IL-2 gene expression and production occur, the IL-2R is expressed, and T cell blastogenesis and proliferation are induced. We have previously found that, although both CD4+ and CD8+ T cell subsets transcribe IL-2, the CD4+ subset appears to be the major producer of IL-2 whereas the CD8+ subset appears to be the major proliferating population when the subsets are separated after activation in vivo. The studies presented here were undertaken to examine the contribution made by the CD4+ subset to lymphocyte proliferation in vivo. Responses to LCMV infection were examined in intact mice and in mice depleted of CD4+ or CD8+ subsets by antibody treatments in vivo. Protocols were such that in vivo treatments with anti-CD4 or anti-CD8 depleted the respective subset by greater than 90%. In situ hybridizations demonstrated that the IL-2 gene was expressed in non-B lymphocytes isolated from either CD4+ cell-depleted or CD8+ cell-depleted mice on day 7 post-infection with LCMV. When placed in culture, however, cells from CD8+ cell-depleted mice produced significantly higher levels of detectable IL-2 than did cells isolated from CD4+ cell-depleted mice on day 7 post-infection. IL-2 was apparently produced in vivo in mice depleted of either CD4+ or CD8+ cells, as expression of the gene for the p55 chain of the IL-2R, IL-2 responsiveness, and lymphocyte proliferation were observed with cells isolated from both sets of mice. Lymphocyte proliferation was shown to be sustained in mice depleted of CD4+ cells in vivo by three criteria: 1) non-B lymphocytes isolated from infected mice depleted of CD4+ cells underwent more DNA synthesis than did those isolated from uninfected mice or from infected mice depleted of CD8+ cells; 2) leukocyte yields were expanded during infection of CD4+ cell-depleted mice; and 3) CD8+ cell numbers were increased during infection of CD4+ cell-depleted mice. The majority of non-B lymphocytes having the characteristics of blast lymphocytes was recovered in the CD8+ populations isolated from infected CD4+ cell-depleted mice. These findings suggest that the requirement for the CD4+ subset to sustain CD8+ lymphocyte proliferation in vivo is limited, and that CD4+ and CD8+ cell types can function independently in many aspects of their responses to viral infections.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Differentiation, T-Lymphocyte , CD8 Antigens , Cell Division , Gene Expression Regulation/immunology , Interleukin-2/biosynthesis , Interleukin-2/genetics , Lymphocyte Depletion , Male , Mice , Mice, Inbred C3H , Receptors, Interleukin-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...