Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Biol ; 448(1): 36-47, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30695685

RESUMEN

Joubert syndrome (JBTS) is a predominantly autosomal recessive neurodevelopmental disorder that presents with characteristic malformations of the cerebellar vermis, superior cerebellar peduncles and midbrain in humans. Accompanying these malformations are a heterogeneous set of clinical symptoms, which frequently include deficits in motor and muscle function, such as hypotonia (low muscle tone) and ataxia (clumsiness). These symptoms are attributed to improper development of the hindbrain, but no direct evidence has been reported linking these in JBTS. Here, we describe muscle developmental defects in a mouse with a targeted deletion of the Abelson helper integration site 1 gene, Ahi1, one of the genes known to cause JBTS in humans. While FVB/NJ Ahi1-/- mice display no gross malformations of the cerebellum, deficits are observed in several measures of motor function, strength, and body development. Specifically, Ahi1-/- mice show delayed physical development, delays in surface reflex righting as neonates, and reductions in grip strength and spontaneous locomotor activity as adults. Additionally, Ahi1-/- mice showed evidence of muscle-specific contributions to this phenotype, such as reductions in 1) myoblast differentiation potential in vitro, 2) muscle desmin expression, and 3) overall muscle mass, myonuclear domain, and muscle fiber cross-sectional area. Together, these data suggest that loss of Ahi1 may cause abnormalities in the differentiation of myoblasts to mature muscle cells. Moreover, Ahi1 loss impacts muscle development directly, outside of any indirect impact of cerebellar malformations, revealing a novel myogenic cause for hypotonia in JBTS.


Asunto(s)
Anomalías Múltiples/embriología , Diferenciación Celular , Cerebelo/anomalías , Anomalías del Ojo/embriología , Enfermedades Renales Quísticas/embriología , Desarrollo de Músculos , Trastornos del Neurodesarrollo/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Retina/anomalías , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Proteínas Adaptadoras del Transporte Vesicular , Animales , Cerebelo/embriología , Cerebelo/patología , Desmina/genética , Desmina/metabolismo , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Locomoción/genética , Ratones , Ratones Noqueados , Fuerza Muscular/genética , Mioblastos/metabolismo , Mioblastos/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Proteínas Proto-Oncogénicas/metabolismo , Reflejo de Enderezamiento/genética , Retina/embriología , Retina/patología
2.
Brain Behav ; 8(6): e00989, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-30106247

RESUMEN

INTRODUCTION: Studying innate sensitivity to ethanol can be an important first step toward understanding alcohol use disorders. In brain, we investigated transcripts, with evidence of miRNA modulation related to a predisposition to the hypnotic effect of ethanol, as measured by loss of righting reflex (LORR). METHODS: Expression of miRNAs (12 samples) and expression of mRNAs (353 samples) in brain were independently analyzed for an association with LORR in mice from the LXS recombinant inbred panel gathered across several small studies. These results were then integrated via a meta-analysis of miRNA-mRNA target pairs identified in miRNA-target interaction databases. RESULTS: We found 112 significant miRNA-mRNA pairs where a large majority of miRNAs and mRNAs were highly interconnected. Most pairs indicated a pattern of increased levels of miRNAs and reduced levels of mRNAs being associated with more alcohol-sensitive strains. For example, CaMKIIn1 was targeted by multiple miRNAs associated with LORR. CAMK2N1 is an inhibitor of CAMK2, which among other functions, phosphorylates, or binds to GABAA and NMDA receptors. CONCLUSIONS: Our results suggest a novel role of miRNA-mediated regulation of an inhibitor of CAMK2 and its downstream targets including the GABAA and NMDA receptors, which have been previously implicated to have a role in ethanol-induced sedation and sensitivity.


Asunto(s)
Alcoholismo/genética , Etanol/farmacología , Hipnóticos y Sedantes/farmacología , MicroARNs/fisiología , Transcripción Genética/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Femenino , Antagonistas de Receptores de GABA-A/farmacología , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Ratones , Ratones Endogámicos , MicroARNs/genética , MicroARNs/metabolismo , ARN Mensajero/metabolismo , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/genética , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/genética , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética
3.
Glia ; 65(12): 2051-2069, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28925029

RESUMEN

The TAM (Tyro3, Axl, and MerTK) family of receptor tyrosine kinases (RTKs) and their ligands, Gas6 and ProS1, are important for innate immune responses and central nervous system (CNS) homeostasis. While only Gas6 directly activates Axl, ProS1 activation of Tyro3/MerTK can indirectly activate Axl through receptor heterodimerization. Therefore, we generated Gas6-/- Axl-/- double knockout (DKO) mice to specifically examine the contribution of this signaling axis while retaining ProS1 signaling through Tyro3 and MerTK. We found that naïve young adult DKO and WT mice have comparable myelination and equal numbers of axons and oligodendrocytes in the corpus callosum. Using the cuprizone model of demyelination/remyelination, transmission electron microscopy revealed extensive axonal swellings containing autophagolysosomes and multivesicular bodies, and fewer myelinated axons in brains of DKO mice at 3-weeks recovery from a 6-week cuprizone diet. Analysis of immunofluorescent staining demonstrated more SMI32+ and APP+ axons and less myelin in the DKO mice. There were no significant differences in the number of GFAP+ astrocytes or Iba1+ microglia/macrophages between the groups of mice. However, at 6-weeks cuprizone and recovery, DKO mice had increased proinflammatory cytokine and altered suppressor of cytokine signaling (SOCS) mRNA expression supporting a role for Gas6-Axl signaling in proinflammatory cytokine suppression. Significant motor deficits in DKO mice relative to WT mice on cuprizone were also observed. These data suggest that Gas6-Axl signaling plays an important role in maintaining axonal integrity and regulating and reducing CNS inflammation that cannot be compensated for by ProS1/Tyro3/MerTK signaling.


Asunto(s)
Axones/patología , Regulación de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Trastornos del Movimiento , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Tirosina Quinasas Receptoras/deficiencia , Remielinización/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Axones/ultraestructura , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Encefalitis/inducido químicamente , Encefalitis/patología , Regulación de la Expresión Génica/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibidores de la Monoaminooxidasa/toxicidad , Trastornos del Movimiento/etiología , Trastornos del Movimiento/genética , Trastornos del Movimiento/patología , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Vaina de Mielina/ultraestructura , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Propiocepción/efectos de los fármacos , Propiocepción/genética , Proteínas Proto-Oncogénicas/genética , Desempeño Psicomotor/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/genética , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética , Remielinización/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tirosina Quinasa del Receptor Axl
4.
Acta Neuropathol ; 133(3): 463-483, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28000031

RESUMEN

Alcohol is a widely consumed drug that can lead to addiction and severe brain damage. However, alcohol is also used as self-medication for psychiatric problems, such as depression, frequently resulting in depression-alcoholism comorbidity. Here, we identify the first molecular mechanism for alcohol use with the goal to self-medicate and ameliorate the behavioral symptoms of a genetically induced innate depression. An induced over-expression of acid sphingomyelinase (ASM), as was observed in depressed patients, enhanced the consumption of alcohol in a mouse model of depression. ASM hyperactivity facilitates the establishment of the conditioned behavioral effects of alcohol, and thus drug memories. Opposite effects on drinking and alcohol reward learning were observed in animals with reduced ASM function. Importantly, free-choice alcohol drinking-but not forced alcohol exposure-reduces depression-like behavior selectively in depressed animals through the normalization of brain ASM activity. No such effects were observed in normal mice. ASM hyperactivity caused sphingolipid and subsequent monoamine transmitter hypo-activity in the brain. Free-choice alcohol drinking restores nucleus accumbens sphingolipid- and monoamine homeostasis selectively in depressed mice. A gene expression analysis suggested strong control of ASM on the expression of genes related to the regulation of pH, ion transmembrane transport, behavioral fear response, neuroprotection and neuropeptide signaling pathways. These findings suggest that the paradoxical antidepressant effects of alcohol in depressed organisms are mediated by ASM and its control of sphingolipid homeostasis. Both emerge as a new treatment target specifically for depression-induced alcoholism.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Etanol/uso terapéutico , Homeostasis/genética , Esfingolípidos/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Animales , Conducta de Elección/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Depresión/genética , Etanol/sangre , Preferencias Alimentarias/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Esfingomielina Fosfodiesterasa/genética , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
5.
Neurobiol Dis ; 96: 271-283, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27693510

RESUMEN

We identified a novel spontaneous mutant mouse showing motor symptoms that are similar to those of the dystonia musculorum (dt) mouse. The observations suggested that the mutant mice inherited the mild dt phenotype as an autosomal recessive trait. Linkage analysis showed that the causative gene was located near D1Mit373 and D1Mit410 microsatellite markers on chromosome 1, which are close to the dystonin (Dst) gene locus. To investigate whether Dst is the causative gene of the novel mutant phenotype, we crossed the mutant with Dst gene trap (DstGt) mice. Compound heterozygotes showed a typical dt phenotype with sensory degeneration and progressive motor symptoms. DNA sequencing analysis identified a nonsense mutation within the spectrin repeats of the plakin domain. The novel mutant allele was named dt23Rbrc. Motor abnormalities in homozygous dt23Rbrc/dt23Rbrc mice are not as severe as homozygous DstGt/DstGt mice. Histological analyses showed abnormal neurofilament (NF) accumulation in the nervous system of homozygous dt23Rbrc/dt23Rbrc mice, which is characteristic of the dt phenotype. We mapped the distribution of abnormal NF-accumulated neurons in the brain and found that they were located specifically in the brainstem, spinal cord, and in regions such as the vestibular nucleus, reticular nucleus, and red nucleus, which are implicated in posture and motor coordination pathways. The quantification of abnormal NF accumulation in the cytoplasm and spheroids (axons) of neurons showed that abnormal NF immunoreactivity was lower in homozygous dt23Rbrc/dt23Rbrc mice than in homozygous DstGt/DstGt mice. Therefore, we have identified a novel hypomorphic allele of dt, which causes histological abnormalities in the central nervous system that may account for the abnormal motor phenotype. This novel spontaneously occurring mutant may become a good model of hereditary sensory and autonomic neuropathy type 6, which is caused by mutations in the human DST gene.


Asunto(s)
Trastornos Distónicos/complicaciones , Trastornos Distónicos/genética , Distonina/genética , Regulación del Desarrollo de la Expresión Génica/genética , Trastornos Heredodegenerativos del Sistema Nervioso/etiología , Repeticiones de Microsatélite/genética , Factores de Edad , Animales , Animales Recién Nacidos , Cromosomas Humanos Par 1/genética , Trastornos Distónicos/patología , Distonina/metabolismo , Potenciales Evocados Motores/genética , Conducta Exploratoria/fisiología , Genotipo , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Trastornos Heredodegenerativos del Sistema Nervioso/patología , Humanos , Filamentos Intermedios/genética , Filamentos Intermedios/metabolismo , Filamentos Intermedios/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Actividad Motora/genética , Neuronas/fisiología , Neuronas/ultraestructura , Reflejo de Enderezamiento/genética , Percepción Espacial/fisiología
6.
Eur J Neurosci ; 40(1): 2311-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24674448

RESUMEN

How external stimuli prevent the onset of sleep has been little studied. This is usually considered to be a non-specific type of phenomenon. However, the hypnotic drug dexmedetomidine, an agonist at α2 adrenergic receptors, has unusual properties that make it useful for investigating this question. Dexmedetomidine is considered to produce an 'arousable' sleep-like state, so that patients or animals given dexmedetomidine become alert following modest stimulation. We hypothesized that it might be more difficult to make mice unconscious with dexmedetomidine if there was a sufficient external stimulus. Employing a motorized rotating cylinder, which provided a continuous and controlled arousal stimulus, we quantitatively measured the ability of such a stimulus to prevent dexmedetomidine loss of righting reflex in two inbred strains of mice (C57BL/6 and 129X1). We found that whereas the C57BL/6 strain required a strong stimulus to prevent dexmedetomidine-induced hypnosis, the 129X1 strain stayed awake even with minimal stimuli. Remarkably, this could be calibrated as a simple threshold trait, i.e. a binary 'yes-no' response, which after crossing the two mouse strains behaved as a dominant-like trait. We carried out a genome-wide linkage analysis on the F2 progeny to determine if the ability of a stimulus to prevent dexmedetomidine hypnosis could be mapped to one or more chromosomal regions. We identified a locus on chromosome 4 with an associated Logarithm of Odds score exceeding the pre-established threshold level. These results show that complex traits, such as the ability of a stimulus to reverse drug-induced hypnosis, may have precise genetic determinants.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Dexmedetomidina/farmacología , Sueño/genética , Vigilia/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Cromosomas de los Mamíferos , Electroencefalografía , Genes Dominantes , Estudio de Asociación del Genoma Completo , Hipnóticos y Sedantes/farmacología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Farmacogenética , Estimulación Física , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética , Reflejo de Enderezamiento/fisiología , Prueba de Desempeño de Rotación con Aceleración Constante , Sueño/efectos de los fármacos , Sueño/fisiología , Especificidad de la Especie , Vigilia/efectos de los fármacos , Vigilia/fisiología
7.
Neuropharmacology ; 77: 177-84, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24067926

RESUMEN

Serotonergic dysfunction has been hypothesized to play an important role in the pathophysiology of alcoholism. However, whether congenital serotonin (5-HT) deficiency leads to increased alcohol consumption or affects ethanol-related behaviors has not been established. Here, we use a transgenic mouse line that expresses a hypofunctional variant of the 5-HT synthesis enzyme, tryptophan hydroxylase 2, to examine the impact of 5-HT deficiency on responses to alcohol. We demonstrate that these 5-HT-deficient transgenic animals (Tph2KI mice) recover their righting reflex more rapidly than wild-type controls following a high dose of ethanol and exhibit blunted locomotor retardation in response to repeated ethanol administration. In addition, compared to WT controls, 5-HT-deficient animals consume significantly more ethanol and exhibit increased preference for ethanol in two-bottle choice tests. Our data also suggest that 5-HT plays a critical role in mediating the effects of ethanol on Akt/GSK3ß signaling in the nucleus accumbens. Overall, our results corroborate previous theories regarding the importance of brain 5-HT levels in mediating responsiveness to alcohol and demonstrate, for the first time, that congenital 5-HT deficiency leads to increased ethanol consumption and decreased sensitivity to the sedative-like effects of ethanol, perhaps in part through modulating Akt/GSK3ß signaling.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Encéfalo/metabolismo , Etanol/administración & dosificación , Serotonina/deficiencia , Triptófano Hidroxilasa/genética , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Encéfalo/efectos de los fármacos , Conducta de Elección/efectos de los fármacos , Conducta de Elección/fisiología , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética , Triptófano Hidroxilasa/metabolismo
8.
Acta Neurochir Suppl ; 118: 307-10, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23564155

RESUMEN

BACKGROUND: Although protein kinase C-γ (PKC-γ) is a target for the effects of volatile anesthetics, the molecular mechanisms of the kinase function remain unclear. We examined the effects of different types of anesthetics on PKC-γ knockout mice, and investigated the dynamics of the kinase in mouse brain. METHODS: We measured the required number of times for loss of righting reflex (rtfLORR) after administration of isoflurane, sevoflurane, and propofol on PKC-γ knockout mice and compared with those of wild-type mice. We also used immunoblotting to investigate the intracellular distribution of PKC-γ and phosphorylated PKC-γ (p-PKC-γ) in brain of wild-type mice anesthetized by these anesthetics. RESULTS: Isoflurane and sevoflurane significantly prolonged the rtfLORRs in PKC-γ knockout mice compared with those in wild-type mice, while no significant difference was observed between knockout and wild-type mice treated with propofol. Examination of the cellular fractions showed that PKC-γ was significantly decreased, whereas p-PKC-γ was significantly increased in the synaptic membrane fraction (P2). There was no significant change in the supernatant fraction (S). In propofol-treated mice, PKC-γ and p-PKC-γ showed no significant changes in P2 or S. CONCLUSION: Our results provide new evidence to support the possibility of the involvement of PKC-γ in the actions of volatile anesthetics.


Asunto(s)
Anestésicos por Inhalación/administración & dosificación , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Reflejo de Enderezamiento/efectos de los fármacos , Animales , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteína Quinasa C/deficiencia , Reflejo de Enderezamiento/genética , Factores de Tiempo
9.
J Pharmacol Exp Ther ; 344(2): 489-500, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23230213

RESUMEN

Ethanol is a widely used drug, yet an understanding of its sites and mechanisms of action remains incomplete. Among the protein targets of ethanol are glycine receptors (GlyRs), which are potentiated by millimolar concentrations of ethanol. In addition, zinc ions also modulate GlyR function, and recent evidence suggests that physiologic concentrations of zinc enhance ethanol potentiation of GlyRs. Here, we first built a homology model of a zinc-bound GlyR using the D80 position as a coordination site for a zinc ion. Next, we investigated in vitro the effects of zinc on ethanol action at recombinant wild-type (WT) and mutant α1 GlyRs containing the D80A substitution, which eliminates zinc potentiation. At D80A GlyRs, the effects of 50 and 200 mM ethanol were reduced as compared with WT receptors. Also, in contrast to what was seen with WT GlyRs, neither adding nor chelating zinc changed the magnitude of ethanol enhancement of mutant D80A receptors. Next, we evaluated the in vivo effects of the D80A substitution by using heterozygous Glra1(D80A) knock-in (KI) mice. The KI mice showed decreased ethanol consumption and preference, and they displayed increased startle responses compared with their WT littermates. Other behavioral tests, including ethanol-induced motor incoordination and strychnine-induced convulsions, revealed no differences between the KI and WT mice. Together, our findings indicate that zinc is critical in determining the effects of ethanol at GlyRs and suggest that zinc binding at the D80 position may be important for mediating some of the behavioral effects of ethanol action at GlyRs.


Asunto(s)
Consumo de Bebidas Alcohólicas , Conducta Animal/efectos de los fármacos , Etanol/farmacología , Mutación Puntual , Receptores de Glicina/genética , Zinc/metabolismo , Estimulación Acústica , Consumo de Bebidas Alcohólicas/genética , Sustitución de Aminoácidos , Animales , Sitios de Unión , Fenómenos Electrofisiológicos , Etanol/administración & dosificación , Femenino , Técnicas de Sustitución del Gen , Homocigoto , Masculino , Ratones , Ratones Mutantes , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Oocitos/metabolismo , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/genética , Prueba de Desempeño de Rotación con Aceleración Constante , Estricnina/farmacología , Transfección , Xenopus laevis , Zinc/farmacología
10.
Alcohol ; 46(5): 463-71, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22572056

RESUMEN

Alcohol and nicotine are often co-used and data from human and animals studies have demonstrated that common genes underlie responses to these two drugs. Recently, the genes that code for the subunits of the nicotinic acetylcholine receptors have been implicated as a common genetic mediator for alcohol and nicotine responses. The mammalian genes that code for the α6 and ß3 subunits of the nicotinic acetylcholine receptor (Chrna6 and Chrnb3, respectively) are located adjacent to each other on human and mouse chromosome 8. These subunits have gained attention as potential regulators of drug behaviors because of their expression in the striatum where they have been shown to modulate dopamine release. Human genetic studies have shown that variation in these genes is associated with alcohol phenotypes. In the current experiments, mice lacking the Chrna6 or Chrnb3 gene were tested for three ethanol behaviors: choice ethanol consumption, ataxia, and sedation. Wildtype (WT), heterozygous (HET), and knockout (KO) mice of each strain went through a standard 2-bottle choice drinking paradigm, the balance beam, and the Loss of Righting Reflex (LORR) paradigm. No genotypic effects on any of the 3 behavioral tasks were observed in Chrnb3 animals. While the Chrna6 gene did not significantly influence ethanol consumption (g/kg) or ataxia, mice lacking the α6 subunit took significantly longer to recover their righting reflex than WT animals. These data provide evidence that receptors containing this subunit modulate the sedative effects of ethanol. Further work examining other models of ethanol consumption and behavioral responses to ethanol is needed to fully characterize the role of these receptor subunits in modulating ethanol responses.


Asunto(s)
Receptores Nicotínicos/fisiología , Consumo de Bebidas Alcohólicas/genética , Animales , Conducta Animal/efectos de los fármacos , Conducta de Elección/efectos de los fármacos , Etanol/farmacología , Femenino , Hipnóticos y Sedantes/farmacología , Masculino , Ratones , Ratones Noqueados , Receptores Nicotínicos/efectos de los fármacos , Reflejo de Enderezamiento/genética
11.
J Pharmacol Exp Ther ; 342(1): 222-31, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22532627

RESUMEN

Thyrotropin-releasing hormone (TRH; pGlu-His-Pro-NH(2)) has multiple, but transient, homeostatic functions in the brain. It is hydrolyzed in vitro by pyroglutamyl peptidase II (PPII), a narrow specificity ectoenzyme with a preferential localization in the brain, but evidence that PPII controls TRH communication in the brain in vivo is scarce. We therefore studied in male Wistar rats the distribution of PPII mRNA in the septum and the consequence of PPII inhibition on the analeptic effect of TRH injected into the medial septum. Twelve to 14% of cell profiles expressed PPII mRNA in the medial septum-diagonal band of Broca; in this region the specific activity of PPII was relatively high. Twenty to 35% of PPII mRNA-labeled profiles were positive for TRH-receptor 1 (TRH-R1) mRNA. The intramedial septum injection of TRH reduced, in a dose-dependent manner, the duration of ethanol-induced loss of righting reflex (LORR). Injection of the PPII inhibitor pGlu-Asn-Pro-7-amido-4-methylcoumarin into the medial septum enhanced the effect of TRH. The injection of a phosphinic TRH analog, a higher-affinity inhibitor of PPII, diminished the duration of LORR by itself. In contrast, the intraseptal injection of pGlu-Asp-Pro-NH(2), a peptide that did not inhibit PPII activity, or an inhibitor of prolyl oligopeptidase did not change the duration of LORR. We conclude that in the medial septum PPII activity may limit TRH action, presumably by reducing the concentration of TRH in the extracellular fluid around cells coexpressing PPII and TRH-R1.


Asunto(s)
Aminopeptidasas/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/farmacología , Ácido Pirrolidona Carboxílico/análogos & derivados , Tabique del Cerebro/efectos de los fármacos , Tabique del Cerebro/enzimología , Hormona Liberadora de Tirotropina/farmacología , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Animales , Masculino , Péptidos/farmacología , Prolil Oligopeptidasas , Ácido Pirrolidona Carboxílico/antagonistas & inhibidores , Ácido Pirrolidona Carboxílico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Tirotropina/genética , Receptores de Hormona Liberadora de Tirotropina/metabolismo , Reflejo de Enderezamiento/genética , Tabique del Cerebro/metabolismo , Serina Endopeptidasas/farmacología
12.
Alcohol Clin Exp Res ; 36(7): 1162-70, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22316249

RESUMEN

BACKGROUND: Mouse lines are being selectively bred in replicate for high blood ethanol concentrations (BECs) achieved after a short period of ethanol (EtOH) drinking early in the circadian dark phase. High Drinking in the Dark-1 (HDID-1) mice were in selected generation S18, and the replicate HDID-2 line in generation S11. METHODS: To determine other traits genetically correlated with high DID, we compared naïve animals from both lines with the unselected, segregating progenitor stock, HS/Npt. Differences between HDID-1 and HS would imply commonality of genetic influences on DID and these traits. RESULTS: HDID-1 mice showed less basal activity, greater EtOH stimulated activity, and greater sensitivity to EtOH-induced foot slips than HS. They showed lesser sensitivity to acute EtOH hypothermia and longer duration loss of righting reflex than HS. HDID-1 and control HS lines did not differ in sensitivity on 2 measures of intoxication, the balance beam and the accelerating rotarod. None of the acute response results could be explained by differences in EtOH metabolism. HDID-2 differed from HS on some, but not all, of the above responses. CONCLUSIONS: These results show that some EtOH responses share common genetic control with reaching high BECs after DID, a finding consistent with other data regarding genetic contributions to EtOH responses.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Cruzamiento , Etanol/administración & dosificación , Animales , Ritmo Circadiano/efectos de los fármacos , Ritmo Circadiano/genética , Etanol/efectos adversos , Femenino , Hipotermia/inducido químicamente , Hipotermia/genética , Masculino , Ratones , Ratones Transgénicos , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética , Especificidad de la Especie
13.
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
14.
Neuropharmacology ; 62(5-6): 1999-2009, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22248636

RESUMEN

Neuropeptide S (NPS) is the endogenous ligand of a previously orphan receptor now named NPSR. In the brain NPS regulates several biological functions including anxiety, arousal, locomotion, food intake, learning and memory, pain and drug abuse. Mice lacking the NPSR gene (NPSR(-/-)) represent an useful tool to investigate the neurobiology of the NPS/NPSR system. NPSR(-/-) mice have been generated in a 129S6/SvEv genetic background. In the present study we generated CD-1 congenic NPSR(+/+) and NPSR(-/-) mice and investigated their phenotype and sensitivity to NPS in various behavioural assays. The phenotype analysis revealed no locomotor differences between NPSR(+/+) and NPSR(-/-) mice. The behaviour of NPSR(+/+) and NPSR(-/-) mice in the righting reflex test was superimposable. No differences were recorded between the two genotypes in the elevated plus maze, open field and stress-induced hyperthermia tests, with the exception of rearing behaviour that was reduced in knockout animals. Moreover the behaviour of NPSR(+/+) and NPSR(-/-) mice in the forced swimming, novel object recognition and formalin assays was similar. The stimulatory effects of NPS in the locomotor activity test and its anxiolytic-like actions in the elevated plus maze and open field assays were evident in NPSR(+/+) but not NPSR(-/-) animals. In conclusion, the present study indicates that the NPS/NPSR system does not tonically control locomotion, sensitivity to diazepam, anxiety, depressive-like behaviours, memory and pain transmission in mice. Furthermore our results clearly show that the product of the NPSR gene represents the mandatory protein for all the NPS biological effects so far described.


Asunto(s)
Conducta Animal/fisiología , Actividad Motora/genética , Receptores Acoplados a Proteínas G/metabolismo , Reflejo de Enderezamiento/genética , Animales , Ansiedad/genética , Depresión/genética , Memoria/fisiología , Ratones , Ratones Noqueados , Dolor/genética , Fenotipo , Receptores Acoplados a Proteínas G/genética
15.
J Pharmacol Exp Ther ; 340(2): 317-29, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22037202

RESUMEN

We used behavioral pharmacology to characterize heterozygous knockin mice with mutations (Q266I or M287L) in the α1 subunit of the glycine receptor (GlyR) (J Pharmacol Exp Ther 340:304-316, 2012). These mutations were designed to reduce (M287L) or eliminate (Q266I) ethanol potentiation of GlyR function. We asked which behavioral effects of ethanol would be reduced more in the Q266I mutant than the M287L and found rotarod ataxia to be the behavior that fulfilled this criterion. Compared with controls, the mutant mice also differed in ethanol consumption, ethanol-stimulated startle response, signs of acute physical dependence, and duration of loss of righting response produced by ethanol, butanol, ketamine, pentobarbital, and flurazepam. Some of these behavioral changes were mimicked in wild-type mice by acute injections of low, subconvulsive doses of strychnine. Both mutants showed increased acoustic startle response and increased sensitivity to strychnine seizures. Thus, in addition to reducing ethanol action on the GlyRs, these mutations reduced glycinergic inhibition, which may also alter sensitivity to GABAergic drugs.


Asunto(s)
Sustitución de Aminoácidos/fisiología , Conducta Animal/fisiología , Mutación/fisiología , Receptores de Glicina/fisiología , 1-Butanol/farmacología , Consumo de Bebidas Alcohólicas/genética , Convulsiones por Abstinencia de Alcohol/diagnóstico , Convulsiones por Abstinencia de Alcohol/genética , Animales , Conducta Animal/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Condicionamiento Clásico , Sinergismo Farmacológico , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Etanol/administración & dosificación , Etanol/metabolismo , Etanol/farmacología , Femenino , Preferencias Alimentarias/efectos de los fármacos , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Destreza Motora/efectos de los fármacos , N-Metilaspartato/farmacología , Nicotina/farmacología , Pentilenotetrazol/farmacología , Quinina/administración & dosificación , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/genética , Prueba de Desempeño de Rotación con Aceleración Constante , Sacarina/administración & dosificación , Convulsiones/inducido químicamente , Convulsiones/diagnóstico , Caracteres Sexuales , Estricnina/farmacología , Percepción del Gusto/genética
16.
Neuroscience ; 171(3): 788-93, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20875840

RESUMEN

Excitatory amino acid transporters (EAAT) uptake extracellular glutamate, the major excitatory neurotransmitter in the brain. EAAT type 3 (EAAT3), the main neuronal EAAT, is expressed widely in the CNS. We have shown that the volatile anesthetic isoflurane increases EAAT3 activity and trafficking to the plasma membrane. Thus, we hypothesize that EAAT3 mediates isoflurane-induced anesthesia. To test this hypothesis, the potency of isoflurane to induce immobility and hypnosis, two major components of general anesthesia, was compared in the CD-1 wild-type mice and EAAT knockout mice that had a CD-1 strain gene background. Hypnosis was assessed by loss of righting reflex in this study. The expression of EAAT1 and EAAT2, two widely expressed EAATs in the CNS, in the cerebral cortex and spinal cord was not different between the EAAT3 knockout mice and wild-type mice. The concentration required for isoflurane to cause immobility to painful stimuli, a response involving primarily reflex loops in the spinal cord, was not changed by EAAT3 knockout. However, the EAAT3 knockout mice were more sensitive to isoflurane-induced hypnotic effects, which may be mediated by hypothalamic sleep neural circuits. Interestingly, the EAAT3 knockout mice did not have an altered sensitivity to the hypnotic effects caused by ketamine, an i.v. anesthetic that is a glutamate receptor antagonist and does not affect EAAT3 activity. These results suggest that EAAT3 modulates the sensitivity of neural circuits to isoflurane. These results, along with our previous findings which suggests that isoflurane increases EAAT3 activity, indicate that EAAT3 may regulate isoflurane-induced behavioral changes, including anesthesia.


Asunto(s)
Transportador 3 de Aminoácidos Excitadores/deficiencia , Transportador 3 de Aminoácidos Excitadores/genética , Isoflurano/farmacología , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética , Anestésicos Disociativos , Anestésicos por Inhalación , Animales , Química Encefálica/efectos de los fármacos , Química Encefálica/genética , Ketamina/farmacología , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA