Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Neuropsychopharmacology ; 48(10): 1446-1454, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37055488

RESUMEN

Drugs that stimulate the trace amine-associated receptor 1 (TAAR1) are under clinical investigation as treatments for several neuropsychiatric disorders. Previous studies in a genetic mouse model of voluntary methamphetamine intake identified TAAR1, expressed by the Taar1 gene, as a critical mediator of aversive methamphetamine effects. Methamphetamine is a TAAR1 agonist, but also has actions at monoamine transporters. Whether exclusive activation of TAAR1 has aversive effects was not known at the time we conducted our studies. Mice were tested for aversive effects of the selective TAAR1 agonist, RO5256390, using taste and place conditioning procedures. Hypothermic and locomotor effects were also examined, based on prior evidence of TAAR1 mediation. Male and female mice of several genetic models were used, including lines selectively bred for high and low methamphetamine drinking, a knock-in line in which a mutant form of Taar1 that codes for a non-functional TAAR1 was replaced by the reference Taar1 allele that codes for functional TAAR1, and their matched control line. RO5256390 had robust aversive, hypothermic and locomotor suppressing effects that were found only in mice with functional TAAR1. Knock-in of the reference Taar1 allele rescued these phenotypes in a genetic model that normally lacks TAAR1 function. Our study provides important data on TAAR1 function in aversive, locomotor, and thermoregulatory effects that are important to consider when developing TAAR1 agonists as therapeutic drugs. Because other drugs can have similar consequences, potential additive effects should be carefully considered as these treatment agents are being developed.


Asunto(s)
Metanfetamina , Ratones , Masculino , Femenino , Animales , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/agonistas
2.
Front Psychiatry ; 12: 725839, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512422

RESUMEN

Sensitivity to rewarding and reinforcing drug effects has a critical role in initial use, but the role of initial aversive drug effects has received less attention. Methamphetamine effects on dopamine re-uptake and efflux are associated with its addiction potential. However, methamphetamine also serves as a substrate for the trace amine-associated receptor 1 (TAAR1). Growing evidence in animal models indicates that increasing TAAR1 function reduces drug self-administration and intake. We previously determined that a non-synonymous single nucleotide polymorphism (SNP) in Taar1 predicts a conformational change in the receptor that has functional consequences. A Taar1 m1J mutant allele existing in DBA/2J mice expresses a non-functional receptor. In comparison to mice that possess one or more copies of the reference Taar1 allele (Taar1 +/+ or Taar1 +/m1J ), mice with the Taar1 m1J/m1J genotype readily consume methamphetamine, express low sensitivity to aversive effects of methamphetamine, and lack sensitivity to acute methamphetamine-induced hypothermia. We used three sets of knock-in and control mice in which one Taar1 allele was exchanged with the alternative allele to determine if other methamphetamine-related traits and an opioid trait are impacted by the same Taar1 SNP proven to affect MA consumption and hypothermia. First, we measured sensitivity to conditioned rewarding and aversive effects of methamphetamine to determine if an impact of the Taar1 SNP on these traits could be proven. Next, we used multiple genetic backgrounds to study the consistency of Taar1 allelic effects on methamphetamine intake and hypothermia. Finally, we studied morphine-induced hypothermia to confirm prior data suggesting that a gene in linkage disequilibrium with Taar1, rather than Taar1, accounts for prior observed differences in sensitivity. We found that a single SNP exchange reduced sensitivity to methamphetamine conditioned reward and increased sensitivity to conditioned aversion. Profound differences in methamphetamine intake and hypothermia consistently corresponded with genotype at the SNP location, with only slight variation in magnitude across genetic backgrounds. Morphine-induced hypothermia was not dependent on Taar1 genotype. Thus, Taar1 genotype and TAAR1 function impact multiple methamphetamine-related effects that likely predict the potential for methamphetamine use. These data support further investigation of their potential roles in risk for methamphetamine addiction and therapeutic development.

3.
Genes Brain Behav ; 20(2): e12667, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32424970

RESUMEN

Trace amine-associated receptor 1 (Taar1) impacts methamphetamine (MA) intake. A mutant allele (Taar1m1J ) derived from the DBA/2J mouse strain codes for a non-functional receptor, and Taar1m1J/m1J mice consume more MA than mice possessing the reference Taar1+ allele. To study the impact of this mutation in a genetically diverse population, heterogeneous stock-collaborative cross (HS-CC) mice, the product of an eight-way cross of standard and wild-derived strains, were tested for MA intake. HS-CC had low MA intake, so an HS-CC by DBA/2J strain F2 intercross was created to transfer the mutant allele onto the diverse background, and used for selective breeding. To study residual variation in MA intake existing in Taar1m1J/m1J mice, selective breeding for higher (MAH) vs lower (MAL) MA intake was initiated from Taar1m1J/m1J F2 individuals; a control line of Taar1+/+ individuals (MAC) was retained. The lines were also examined for MA-induced locomotor and thermal responses, and fluid and tastant consumption. Taar1m1J/m1J F2 mice consumed significantly more MA than Taar1+/+ F2 mice. Response to selection was significant by generation 2 and there were corresponding differences in fluid consumed. Fluid consumption was not different in non-MA drinking studies. Taar1m1J/m1J genotype (MAL or MAH vs MAC mice) was associated with heighted MA locomotor and reduced hypothermic responses. MAL mice exhibited greater sensitization than MAH mice, but the selected lines did not consistently differ for thermal or tastant phenotypes. Residual variation among high-risk Taar1m1J/m1J mice appears to involve mechanisms associated with neuroadaptation to MA, but not sensitivity to hypothermic effects of MA.


Asunto(s)
Trastornos Relacionados con Anfetaminas/genética , Genes Modificadores , Receptores Acoplados a Proteínas G/genética , Selección Artificial , Trastornos Relacionados con Anfetaminas/fisiopatología , Animales , Temperatura Corporal , Conducta Alimentaria , Femenino , Hibridación Genética , Locomoción , Masculino , Metanfetamina/administración & dosificación , Metanfetamina/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Fenotipo
4.
Elife ; 82019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31274109

RESUMEN

We identified a locus on mouse chromosome 10 that accounts for 60% of the genetic variance in methamphetamine intake in mice selectively bred for high versus low methamphetamine consumption. We nominated the trace amine-associated receptor 1 gene, Taar1, as the strongest candidate and identified regulation of the mu-opioid receptor 1 gene, Oprm1, as another contributor. This study exploited CRISPR-Cas9 to test the causal role of Taar1 in methamphetamine intake and a genetically-associated thermal response to methamphetamine. The methamphetamine-related traits were rescued, converting them to levels found in methamphetamine-avoiding animals. We used a family of recombinant inbred mouse strains for interval mapping and to examine independent and epistatic effects of Taar1 and Oprm1. Both methamphetamine intake and the thermal response mapped to Taar1 and the independent effect of Taar1 was dependent on genotype at Oprm1. Our findings encourage investigation of the contribution of Taar1 and Oprm1 variants to human methamphetamine addiction.


Asunto(s)
Variación Genética , Metanfetamina/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Opioides mu/metabolismo , Animales , Secuencia de Bases , Temperatura Corporal , Cromosomas de los Mamíferos/genética , Femenino , Genotipo , Hipotermia/genética , Masculino , Ratones , Sitios de Carácter Cuantitativo/genética , Receptores Acoplados a Proteínas G/metabolismo
5.
Brain Sci ; 9(7)2019 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-31262025

RESUMEN

Transcriptome profiling can broadly characterize drug effects and risk for addiction in the absence of drug exposure. Modern large-scale molecular methods, including RNA-sequencing (RNA-Seq), have been extensively applied to alcohol-related disease traits, but rarely to risk for methamphetamine (MA) addiction. We used RNA-Seq data from selectively bred mice with high or low risk for voluntary MA intake to construct coexpression and cosplicing networks for differential risk. Three brain reward circuitry regions were explored, the nucleus accumbens (NAc), prefrontal cortex (PFC), and ventral midbrain (VMB). With respect to differential gene expression and wiring, the VMB was more strongly affected than either the PFC or NAc. Coexpression network connectivity was higher in the low MA drinking line than in the high MA drinking line in the VMB, oppositely affected in the NAc, and little impacted in the PFC. Gene modules protected from the effects of selection may help to eliminate certain mechanisms from significant involvement in risk for MA intake. One such module was enriched in genes with dopamine-associated annotations. Overall, the data suggest that mitochondrial function and glutamate-mediated synaptic plasticity have key roles in the outcomes of selective breeding for high versus low levels of MA intake.

6.
Front Pharmacol ; 8: 993, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29403379

RESUMEN

Major gene effects on traits associated with substance use disorders are rare. Previous findings in methamphetamine drinking (MADR) lines of mice, bred for high or low voluntary MA intake, and in null mutants demonstrate a major impact of the trace amine-associated receptor 1 (Taar1) gene on a triad of MA-related traits: MA consumption, MA-induced conditioned taste aversion and MA-induced hypothermia. While inbred strains are fundamentally genetically stable, rare spontaneous mutations can become fixed and result in new or aberrant phenotypes. A single nucleotide polymorphism in Taar1 that encodes a missense proline to threonine mutation in the second transmembrane domain (Taar1m1J ) has been identified in the DBA/2J strain. MA is an agonist at this receptor, but the receptor produced by Taar1m1J does not respond to MA or endogenous ligands. In the present study, we used progeny of the C57BL/6J × DBA/2J F2 cross, the MADR lines, C57BL/6J × DBA/2J recombinant inbred strains, and DBA/2 mice sourced from four vendors to further examine Taar1-MA phenotype relations and to define the chronology of the fixation of the Taar1m1J mutation. Mice homozygous for Taar1m1J were found at high frequency early in selection for high MA intake in multiple replicates of the high MADR line, whereas Taar1m1J homozygotes were absent in the low MADR line. The homozygous Taar1m1J genotype is causally linked to increased MA intake, reduced MA-induced conditioned taste aversion, and reduced MA-induced hypothermia across models. Genotype-phenotype correlations range from 0.68 to 0.96. This Taar1 polymorphism exists in DBA/2J mice sourced directly from The Jackson Laboratory, but not DBA/2 mice sourced from Charles River (DBA/2NCrl), Envigo (formerly Harlan Sprague Dawley; DBA/2NHsd) or Taconic (DBA/2NTac). By genotyping archived samples from The Jackson Laboratory, we have determined that this mutation arose in 2001-2003. Our data strengthen the conclusion that the mutant Taar1m1J allele, which codes for a non-functional receptor protein, increases risk for multiple MA-related traits, including MA intake, in homozygous Taar1m1J individuals.

7.
Neuropsychopharmacology ; 42(7): 1539-1547, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27976681

RESUMEN

The psychostimulants amphetamine (AMPH) and methamphetamine (MA) are widely abused illicit drugs. Here we show that both psychostimulants acutely increase NMDA receptor (NMDAR)-mediated synaptic currents and decrease AMPA receptor (AMPAR)/NMDAR ratios in midbrain dopamine neurons. The potentiation depends on the transport of AMPH into the cell by the dopamine transporter. NMDAR-GluN2B receptor inhibitors, ifenprodil, RO 25-6981, and RO 04-5595, inhibit the potentiation without affecting basal-evoked NMDA currents, indicating that NMDAR-GluN2B receptors are activated by AMPH. A selective peptide inhibitor of AMPH-dependent trafficking of the neuronal excitatory amino acid transporter 3 (EAAT3) blocks potentiation, suggesting that EAAT3 internalization increases extracellular glutamate concentrations and activates GluN2B-containing NMDARs. Experiments with the use-dependent NMDAR blocker, MK-801, indicate that potentiated NMDARs reside on the plasma membrane and are not inserted de novo. In behavioral studies, GluN2B inhibitors reduce MA-mediated locomotor activity, without affecting basal activity. These results reveal an important interaction between dopamine and glutamatergic signaling in midbrain dopamine neurons in response to acute administration of psychostimulants.


Asunto(s)
Anfetamina/farmacología , Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/metabolismo , Metanfetamina/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Potenciales Sinápticos/efectos de los fármacos , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Mesencéfalo/efectos de los fármacos , Ratones , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Potenciales Sinápticos/fisiología
8.
Front Neurosci ; 10: 493, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27853417

RESUMEN

Binge/crash cycles of methamphetamine (MA) use are frequently reported by individuals suffering from MA use disorders. A MA binge is self-reported as multiple daily doses that commonly accumulate to 800 mg/day (~10 mg/kg/day for a 170 pound human). A genetic animal model with a similar vulnerability to binge-level MA intake is missing. We used selectively bred MA high drinking (MAHDR) and low drinking (MALDR) mouse lines to determine whether several procedural variations would result in binge-level MA intake. Data were also collected in two progenitor populations of the MA drinking lines, the DBA/2J (D2) strain and the F2 cross of the D2 and C57BL/6J strains. The impact of 3 factors was examined: (1) concentration of MA in the two-bottle choice procedure used for selective breeding; (2) ratio of bottles containing MA vs. water, and (3) length of the withdrawal (or abstinence) period between MA drinking sessions. When MA concentration was progressively increased every 4 days in 20 mg/l amounts from 20 to 140 mg/l, maximum intake in MALDR mice was 1.1 mg/kg, whereas MAHDR mice consumed as much as 14.6 mg/kg. When these concentrations were tested in a multiple bottle choice procedure, the highest ratio of MA to water bottles (3:1) was associated with escalated MA intake of up to 29.1 mg/kg in MAHDR mice and 12.0 mg/kg in F2 mice; MALDR mice did not show a ratio-dependent escalation in MA intake. Finally, MAHDR and D2 mice were offered 3 bottles of MA vs. water at increasing concentrations from 20 to 80 mg/l, and tested under an intermittent 6-h withdrawal period, which was lengthened to 30 h (D2 mice) or to 30 or 78 h (MAHDR). D2 and MAHDR mice initially consumed similar amounts of 14-16 mg/kg MA, but D2 mice reduced their MA intake 3-fold after introduction of 30-h abstinence periods, whereas MAHDR mice retained their high level of intake regardless of withdrawal period. MAHDR mice provide a genetic model of binge-level MA intake appropriate for the study of associated MA-induced neurobiological changes and pharmaceutical treatments.

9.
Expert Opin Drug Discov ; 9(11): 1307-17, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25195620

RESUMEN

INTRODUCTION: There is increasing evidence encouraging the development of drugs that positively modulate the γ-aminobutyric acid type B (GABA(B)) receptor for combating addiction. Compounds that target GABA(B) receptors are unique as anti-abuse therapies because of their impact against multiple addictive drugs. AREAS COVERED: The authors present the basic information concerning the drug actions of GABA and GABA(B) receptor orthosteric agonists and positive allosteric modulators (PAM). Furthermore, they discuss several recent excellent reviews and newer results pertaining to GABA(B) receptor drug effects on responses to and self-administration of: alcohol (ethanol), nicotine, cocaine, (meth)amphetamine, and opioids. Preclinical and clinical data are considered. EXPERT OPINION: Clinical data exist only for baclofen and mostly for alcohol use disorders. Additional trials are needed, but effects are promising. Whether PAMs, given alone or in combination with a direct GABA(B) receptor agonist, will be clinically effective and have fewer side effects requires investigation. The sedative effects of baclofen, a Food and Drug Administration (FDA)-approved drug, become less severe over time. Based on existing data, baclofen is well-tolerated. However, genetic and physiological differences are likely to contribute to individual responses to different therapeutic agents. The more immediate development of baclofen as a therapeutic for alcohol use disorders may be of significant benefit to some individuals.


Asunto(s)
Receptores de GABA-B/metabolismo , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Animales , Baclofeno/farmacología , Descubrimiento de Drogas , Humanos , Trastornos Relacionados con Sustancias/metabolismo
10.
Mamm Genome ; 24(11-12): 446-58, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24217691

RESUMEN

Lines of mice were created by selective breeding for the purpose of identifying genetic mechanisms that influence the magnitude of the selected trait and to explore genetic correlations for additional traits thought to be influenced by shared mechanisms. DNA samples from high and low methamphetamine-drinking (MADR) and high and low methamphetamine-sensitization lines were used for quantitative trait locus (QTL) mapping. Significant additive genetic correlations between the two traits indicated a common genetic influence, and a QTL on chromosome X was detected for both traits, suggesting one source of this commonality. For MADR mice, a QTL on chromosome 10 accounted for more than 50 % of the genetic variance in that trait. Microarray gene expression analyses were performed for three brain regions for methamphetamine-naïve MADR line mice: nucleus accumbens, prefrontal cortex, and ventral midbrain. Many of the genes that were differentially expressed between the high and low MADR lines were shared in common across the three brain regions. A gene network highly enriched in transcription factor genes was identified as being relevant to genetically determined differences in methamphetamine intake. When the mu opioid receptor gene (Oprm1), located on chromosome 10 in the QTL region, was added to this top-ranked transcription factor network, it became a hub in the network. These data are consistent with previously published findings of opioid response and intake differences between the MADR lines and suggest that Oprm1, or a gene that impacts activity of the opioid system, plays a role in genetically determined differences in methamphetamine intake.


Asunto(s)
Metanfetamina/metabolismo , Trastornos Relacionados con Sustancias/genética , Animales , Encéfalo/metabolismo , Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Ratones , Sitios de Carácter Cuantitativo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Trastornos Relacionados con Sustancias/metabolismo
11.
Behav Brain Res ; 256: 420-7, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23994231

RESUMEN

Genetic factors significantly influence addiction-related phenotypes. This is supported by the successful bidirectional selective breeding of two replicate sets of mouse lines for amount of methamphetamine consumed. Some of the same genetic factors that influence methamphetamine consumption have been previously found also to influence sensitivity to the conditioned rewarding and aversive effects of methamphetamine. The goal of the current studies was to determine if some of the same genetic factors influence sensitivity to the conditioned rewarding and aversive effects of cocaine. Cocaine conditioned reward was examined in methamphetamine high drinking and low drinking line mice using a conditioned place preference procedure and cocaine conditioned aversion was measured using a conditioned taste aversion procedure. In addition, a general sensitivity measure, locomotor stimulant response to cocaine, was assessed in these lines; previous data indicated no difference between the selected lines in sensitivity to methamphetamine-induced stimulation. In contrast to robust differences for methamphetamine, the methamphetamine high and low drinking lines did not differ in sensitivity to either the rewarding or aversive effects of cocaine. They also exhibited comparable sensitivity to cocaine-induced locomotor stimulation. These data suggest that the genetic factors that influence sensitivity to the conditioned rewarding and aversive effects of methamphetamine in these lines of mice do not influence sensitivity to these effects of cocaine. Thus, different genetic factors may influence risk for methamphetamine versus cocaine use.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Cocaína/farmacología , Metanfetamina/farmacología , Actividad Motora/efectos de los fármacos , Recompensa , Animales , Aprendizaje por Asociación/efectos de los fármacos , Aprendizaje por Asociación/fisiología , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Actividad Motora/genética , Especificidad de la Especie
12.
J Pharmacol Exp Ther ; 341(2): 455-63, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22333290

RESUMEN

Neuroadaptations underlying sensitization to drugs of abuse seem to influence compulsive drug pursuit and relapse associated with addiction. Our previous data support a role for the corticotropin-releasing factor (CRF) type-1 receptor (CRF1) in ethanol (EtOH)-induced psychomotor sensitization. CRF1 is endogenously activated by CRF and urocortin-1. Because genetic deletion of urocortin-1 did not affect EtOH sensitization, we hypothesized that CRF is the important ligand underlying EtOH sensitization. To test this hypothesis, we used heterozygous and homozygous knockout (KO) mice, which lack one or both copies of the gene coding for CRF, and their respective wild-type controls. EtOH sensitization was normal in heterozygous, but absent in homozygous, CRF KO mice. Corticosterone (CORT) levels were drastically reduced only in CRF KO mice. Because CRF/CRF1 initiate EtOH-induced activation of the hypothalamic-pituitary-adrenal axis, we investigated CORT effects on EtOH sensitization. The CORT synthesis inhibitor metyrapone prevented the acquisition, but not the expression, of EtOH sensitization. Exogenous CORT administration sensitized the locomotor response to a subsequent EtOH challenge; we observed, however, that the exogenous CORT levels necessary to induce sensitization to EtOH were significantly higher than those produced by EtOH treatment. Therefore, participation of CORT seems to be necessary, but not sufficient, to explain the role of CRF/CRF1 in the acquisition of sensitization to EtOH. Extra-hypothalamic CRF/CRF1 mechanisms are suggested to be involved in the expression of EtOH sensitization. The present results are consistent with current theories proposing a key role for CRF and CRF1 in drug-induced neuroplasticity, dependence, and addictive behavior.


Asunto(s)
Conducta Animal/efectos de los fármacos , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Etanol/farmacología , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Animales , Conducta Adictiva/genética , Conducta Adictiva/metabolismo , Hormona Liberadora de Corticotropina/sangre , Hormona Liberadora de Corticotropina/genética , Femenino , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Metirapona/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Trastornos Psicomotores/genética , Trastornos Psicomotores/metabolismo , Receptores de Hormona Liberadora de Corticotropina/genética , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética , Urocortinas/genética , Urocortinas/metabolismo
13.
Psychopharmacology (Berl) ; 220(4): 719-30, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21983918

RESUMEN

RATIONALE: Sensitivity to the stimulant and rewarding effects of alcohol may be genetically correlated traits that predispose individuals to develop an alcohol use disorder. OBJECTIVE: This study aimed to examine the effects of alcohol and cocaine on intracranial self-stimulation (ICSS) in FAST and SLOW mice, which were selectively bred for extremes in alcohol stimulation. METHODS: Male FAST and SLOW mice were conditioned to respond for reinforcement by direct electrical stimulation of the medial forebrain bundle (i.e., brain stimulation reward). ICSS responses were determined immediately before and after oral gavage with water or alcohol (0.3-2.4 g/kg) or intraperitoneal injection with saline or cocaine (1.0-30.0 mg/kg). In separate FAST and SLOW mice, the locomotor effects of these treatments were measured in activity chambers. RESULTS: Alcohol dose-dependently lowered the threshold for self-stimulation (θ (0)) and the frequency that maintained 50% of maximal responding (EF50) in FAST mice but did not significantly affect these parameters in SLOW mice. The largest effects of alcohol were after the 1.7- and 2.4-g/kg doses and were about 40% compared to water injection. Alcohol did not affect MAX response rates, but dose-dependently stimulated locomotor activity in FAST mice. Cocaine lowered thresholds equally in FAST and SLOW mice, although cocaine-stimulated locomotor activity was higher in the FAST than in the SLOW mice. CONCLUSIONS: Selective breeding for alcohol locomotor stimulation also renders the mice more sensitive to the effects of alcohol, but not cocaine, on ICSS.


Asunto(s)
Cocaína/administración & dosificación , Etanol/administración & dosificación , Haz Prosencefálico Medial/efectos de los fármacos , Autoestimulación/efectos de los fármacos , Animales , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica/métodos , Femenino , Masculino , Haz Prosencefálico Medial/fisiología , Ratones , Autoestimulación/fisiología , Especificidad de la Especie
14.
Psychopharmacology (Berl) ; 218(1): 169-77, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21487655

RESUMEN

RATIONALE: Exposure to stressors promotes ethanol (EtOH) consumption and enhances drug craving during abstinence. Corticotropin-releasing factor (CRF), and in particular, CRF actions via type 1 CRF receptors (CRF(1)) are critical in behavioral responses to stressors. CRF(1) play a role in EtOH-induced behavioral neuroadaptation, in binge-like EtOH consumption, and in heightened EtOH consumption in dependent animals. OBJECTIVES: We investigated the involvement of CRF(1) in swim-stress-induced changes in EtOH consumption and in baseline consumption as a function of EtOH concentration. The role of CRF(2) in adapting to effects of the stressor was also examined. METHODS: Wild-type mice and knockout mice lacking CRF(1) were tested for two-bottle choice EtOH consumption at concentrations of 3-20%. Also, intake of 10% EtOH was examined in wild-type mice and knockout mice lacking CRF(1), or lacking both CRF(1) and CRF(2), before and after acute or repeated swim stress exposures. RESULTS: EtOH intake was reduced in CRF(1) compared with wild-type mice when presented at a concentration of 20% but not when presented at lower concentrations. No genotype-dependent effects were found for saccharin or quinine drinking. Acute swim stress had no effect, but repeated swim stress resulted in higher levels of EtOH consumption in wild-type mice, compared with both types of knockout mice. Stress effects on EtOH drinking were longer lasting in double knockout mice. CONCLUSIONS: These data suggest a prominent role of CRF(1) in stressor-induced changes in EtOH consumption, with involvement of CRF(2) in recovery from stressor effects.


Asunto(s)
Etanol/administración & dosificación , Receptores de Hormona Liberadora de Corticotropina/genética , Estrés Psicológico/complicaciones , Consumo de Bebidas Alcohólicas/epidemiología , Consumo de Bebidas Alcohólicas/genética , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quinina/administración & dosificación , Sacarina/administración & dosificación , Estrés Psicológico/genética , Natación , Tiempo
15.
Psychopharmacology (Berl) ; 214(4): 791-804, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21088960

RESUMEN

RATIONALE: Genetically determined differences in susceptibility to drug-induced sensitization could be related to risk for drug consumption. OBJECTIVES: Studies were performed to determine whether selective breeding could be used to create lines of mice with different magnitudes of locomotor sensitization to methamphetamine (MA). MA sensitization (MASENS) lines were also examined for genetically correlated responses to MA. METHODS: Beginning with the F2 cross of C57BL/6J and DBA/2J strains, mice were tested for locomotor sensitization to repeated injections of 1 mg/kg MA and bred based on magnitude of sensitization. Five selected offspring generations were tested. All generations were also tested for MA consumption, and some were tested for dose-dependent locomotor-stimulant responses to MA, consumption of saccharin, quinine, and potassium chloride as a measure of taste sensitivity, and MA clearance after acute and repeated MA. RESULTS: Selective breeding resulted in creation of two lines [MA high sensitization (MAHSENS) and MA low sensitization (MALSENS)] that differed in magnitude of MA-induced sensitization. Initially, greater MA consumption in MAHSENS mice reversed over the course of selection so that MALSENS mice consumed more MA. MAHSENS mice exhibited greater sensitivity to the acute stimulant effects of MA, but there were no significant differences between the lines in MA clearance from blood. CONCLUSIONS: Genetic factors influence magnitude of MA-induced locomotor sensitization and some of the genes involved in magnitude of this response also influence MA sensitivity and consumption. Genetic factors leading to greater MA-induced sensitization may serve a protective role against high levels of MA consumption.


Asunto(s)
Trastornos Relacionados con Anfetaminas/genética , Conducta Animal/efectos de los fármacos , Cruzamiento/métodos , Estimulantes del Sistema Nervioso Central/farmacología , Predisposición Genética a la Enfermedad/genética , Metanfetamina/farmacología , Selección Genética , Animales , Estimulantes del Sistema Nervioso Central/administración & dosificación , Relación Dosis-Respuesta a Droga , Metanfetamina/administración & dosificación , Ratones , Ratones Endogámicos , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Conducta Estereotipada/efectos de los fármacos
16.
Mamm Genome ; 21(1-2): 39-51, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20033183

RESUMEN

Excessive alcohol (ethanol) consumption is the hallmark of alcohol use disorders. The F1 hybrid cross between the C57BL/6J (B6) and FVB/NJ (FVB) inbred mouse strains consumes more ethanol than either progenitor strain. The purpose of this study was to utilize ethanol-drinking data and genetic information to map genes that result in overdominant (or heterotic) ethanol drinking. About 600 B6 x FVB F2 mice, half of each sex, were tested for ethanol intake and preference in a 24-h, two-bottle water versus ethanol choice procedure, with ascending ethanol concentrations. They were then tested for ethanol intake in a Drinking in the Dark (DID) procedure, first when there was no water choice and then when ethanol was offered versus water. DNA samples were obtained and genome-wide QTL analyses were performed to search for single QTLs (both additive and dominance effects) and interactions between pairs of QTLs, or epistasis. On average, F2 mice consumed excessive amounts of ethanol in the 24-h choice procedure, consistent with high levels of consumption seen in the F1 cross. Consumption in the DID procedure was similar or higher than amounts reported previously for the B6 progenitor. QTLs resulting in heightened consumption in heterozygous compared to homozygous animals were found on Chrs 11, 15, and 16 for 24-h choice 30% ethanol consumption, and on Chr 11 for DID. No evidence was found for epistasis between any pair of significant or suggestive QTLs. This indicates that the hybrid overdominance is due to intralocus interactions at the level of individual QTL.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Alcoholismo/genética , Sitios Genéticos/fisiología , Animales , Conducta Animal , Conducta de Elección/fisiología , Mapeo Cromosómico , Cruzamientos Genéticos , Oscuridad , Epistasis Genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Fenotipo , Sitios de Carácter Cuantitativo
17.
Proc Natl Acad Sci U S A ; 105(26): 9070-5, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18591672

RESUMEN

A common expression of neuroadaptations induced by repeated exposure to addictive drugs is a persistent sensitized behavioral response to their stimulant properties. Neuroplasticity underlying drug-induced sensitization has been proposed to explain compulsive drug pursuit and consumption characteristic of addiction. The hypothalamic-pituitary-adrenal (HPA) axis-activating neuropeptide, corticotropin-releasing factor (CRF), may be the keystone in drug-induced neuroadaptation. Corticosterone-activated glucocorticoid receptors (GRs) mediate the development of sensitization to ethanol (EtOH), implicating the HPA axis in this process. EtOH-induced increases in corticosterone require CRF activation of CRF1 receptors. We posited that CRF1 signaling pathways are crucial for EtOH-induced sensitization. We demonstrate that mice lacking CRF1 receptors do not show psychomotor sensitization to EtOH, a phenomenon that was also absent in CRF1 + 2 receptor double-knockout mice. Deletion of CRF2 receptors alone did not prevent sensitization. A blunted endocrine response to EtOH was found only in the genotypes showing no sensitization. The CRF1 receptor antagonist CP-154,526 attenuated the acquisition and prevented the expression of EtOH-induced psychomotor sensitization. Because CRF1 receptors are also activated by urocortin-1 (Ucn1), we tested Ucn1 knockout mice for EtOH sensitization and found normal sensitization in this genotype. Finally, we show that the GR antagonist mifepristone does not block the expression of EtOH sensitization. CRF and CRF1 receptors, therefore, are involved in the neurobiological adaptations that underlie the development and expression of psychomotor sensitization to EtOH. A CRF/CRF1-mediated mechanism involving the HPA axis is proposed for acquisition, whereas an extrahypothalamic CRF/CRF1 participation is suggested for expression of sensitization to EtOH.


Asunto(s)
Adaptación Biológica/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Etanol/farmacología , Fenómenos Fisiológicos del Sistema Nervioso/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Hormona Adrenocorticotrópica/sangre , Animales , Etanol/administración & dosificación , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mifepristona/farmacología , Desempeño Psicomotor/efectos de los fármacos , Pirimidinas/farmacología , Pirroles/farmacología , Urocortinas/metabolismo
18.
Behav Neurosci ; 120(6): 1356-66, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17201481

RESUMEN

Acute drug stimulation has been proposed to be an endophenotype for drug abuse. The authors previously reported the short-term selective breeding of lines of mice for low (LMACT) and high (HMACT) stimulation to methamphetamine (MA). These mice were used to examine whether common genes influence the locomotor response to MA and ethanol. Additionally, the authors tested these mice for ethanol drinking, locomotor sensitization, and clearance. LMACT mice were less stimulated by ethanol and consumed more ethanol than HMACT mice, but the lines did not differ in ethanol-induced sensitization. A small difference in ethanol clearance rate (0.1 mg/ml/h) likely had little impact on behavior. Some common genes may influence the locomotor response to MA and ethanol, as well as ethanol drinking.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Etanol/farmacología , Metanfetamina/farmacología , Actividad Motora/efectos de los fármacos , Selección Genética , Consumo de Bebidas Alcohólicas , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Depresores del Sistema Nervioso Central/sangre , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Etanol/sangre , Ratones , Ratones Endogámicos C57BL , Actividad Motora/genética
19.
Dev Dyn ; 234(1): 74-89, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16059910

RESUMEN

To gain insight into the coordination of gene expression profiles during forelimb and hindlimb differentiation, a transcriptome analysis of mouse embryonic autopod tissues was performed using Affymetrix Murine Gene Chips (MOE-430). Forty-four transcripts with expression differences higher than 2-fold (T test, P < or = 0.05) were detected between forelimb and hindlimb tissues including 38 new transcripts such as Rdh10, Frzb, Tbx18, and Hip that exhibit differential limb expression. A comparison of gene expression profiles in the forelimb, hindlimb, and brain revealed 24 limb-signature genes whose expression was significantly enriched in limb autopod versus brain tissue (fold change >2, P < or = 0.05). Interestingly, the genes exhibiting enrichment in the developing autopod also segregated into significant fore- and hindlimb-specific clusters (P < or = 0.05) suggesting that by E 12.5, unique gene combinations are being used during the differentiation of each autopod type.


Asunto(s)
Miembro Anterior/embriología , Miembro Posterior/embriología , ARN/metabolismo , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Diferenciación Celular , Miembro Anterior/citología , Miembro Anterior/metabolismo , Perfilación de la Expresión Génica , Miembro Posterior/citología , Miembro Posterior/metabolismo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Tiempo , Tretinoina/metabolismo
20.
Mamm Genome ; 16(5): 291-305, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-16104378

RESUMEN

In an effort to identify genes that may be important for drug-abuse liability, we mapped behavioral quantitative trait loci (bQTL) for sensitivity to the locomotor stimulant effect of methamphetamine (MA) using two mouse lines that were selectively bred for high MA-induced activity (HMACT) or low MA-induced activity (LMACT). We then examined gene expression differences between these lines in the nucleus accumbens, using 20 U74Av2 Affymetrix microarrays and quantitative polymerase chain reaction (qPCR). Expression differences were detected for several genes, including Casein Kinase 1 Epsilon (Csnkle), glutamate receptor, ionotropic, AMPA1 (GluR1), GABA B1 receptor (Gabbr1), and dopamine- and cAMP-regulated phosphoprotein of 32 kDa (Darpp-32). We used the www.WebQTL.org database to identify QTL that regulate the expression of the genes identified by the microarrays (expression QTL; eQTL). This approach identified an eQTL for Csnkle on Chromosome 15 (LOD = 3.8) that comapped with a bQTL for the MA stimulation phenotype (LOD = 4.5), suggesting that a single allele may cause both traits. The chromosomal region containing this QTL has previously been associated with sensitivity to the stimulant effects of cocaine. These results suggest that selection was associated with (and likely caused) altered gene expression that is partially attributable to different frequencies of gene expression polymorphisms. Combining classical genetics with analysis of whole-genome gene expression and bioinformatic resources provides a powerful method for provisionally identifying genes that influence complex traits. The identified genes provide excellent candidates for future hypothesis-driven studies, translational genetic studies, and pharmacological interventions.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Metanfetamina/farmacología , Ratones Endogámicos C57BL/genética , Ratones Endogámicos DBA/genética , Animales , Mapeo Cromosómico , Cruzamientos Genéticos , Resistencia a Medicamentos/genética , Femenino , Marcadores Genéticos , Genotipo , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Sitios de Carácter Cuantitativo , ARN/genética , ARN/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...