Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Behav Brain Res ; 370: 111943, 2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31095992

RESUMEN

Adolescent's consumption of caffeine and caffeinated beverage is increasing, yet little is known about the consequences of chronic caffeine exposure during the critical development period of adolescence. In the present study, we investigated the effect of beginning chronic caffeine consumption in adolescence on locomotor, mood, sensorimotor gating, and reward seeking behaviors through adolescence and in adulthood. During the light cycle, caffeine exposed mice exhibited hypoactivity in a novel open-field box and increased anxiety-like and depressive-like behaviors, while maintaining normal home cage locomotor activity. In contrast, during the dark cycle caffeine exposed mice displayed normal locomotor activity in a novel open-field box with hyperactive home cage activity. Interestingly, we found that caffeine exposed mice also showed enhanced prepulse inhibition during the light cycle whereas they displayed a deficit of prepulse inhibition during the dark cycle. Reward seeking for sucrose was higher in caffeine exposed than control mice during the light cycle. Additionally, when granted 24 -h access to ethanol as adults, caffeine exposed mice consumed more ethanol in the absence of acute caffeine use. Altogether, mice that consumed chronic caffeine beginning in adolescence had increased reward seeking and exhibited a circadian-dependent pattern of mood fluctuations in adulthood.


Asunto(s)
Afecto/efectos de los fármacos , Cafeína/farmacología , Motivación/efectos de los fármacos , Factores de Edad , Animales , Ansiedad/metabolismo , Conducta Animal/efectos de los fármacos , Depresión/metabolismo , Locomoción/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Recompensa , Filtrado Sensorial/efectos de los fármacos
2.
Proteomics ; 18(7): e1700417, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29437267

RESUMEN

Acamprosate is an FDA-approved medication for the treatment of alcoholism that is unfortunately only effective in certain patients. Although acamprosate is known to stabilize the hyper-glutamatergic state in alcoholism, pharmacological mechanisms of action in brain tissue remains unknown. To investigate the mechanism of acamprosate efficacy, the authors employ a pharmacoproteomics approach using an animal model of alcoholism, type 1 equilibrative nucleoside transporter (ENT1) null mice. The results demonstrate that acamprosate treatment significantly decreased both ethanol drinking and preference in ENT1 null mice compared to that of wild-type mice. Then, to elucidate acamprosate efficacy mechanism in ENT1 null mice, the authors utilize label-free quantification proteomics comparing both genotype and acamprosate treatment effects in the nucleus accumbens (NAc). A total of 1040 protein expression changes are identified in the NAc among 3634 total proteins detected. The proteomics and Western blot result demonstrate that acamprosate treatment decreased EAAT expression implicating stabilization of the hyper-glutamatergic condition in ENT1 null mice. Pathway analysis suggests that acamprosate treatment in ENT1 null mice seems to rescue glutamate toxicity through restoring of RTN4 and NF-κB medicated neuroimmune signaling compared to wild-type mice. Overall, pharmacoproteomics approaches suggest that neuroimmune restoration is a potential efficacy mechanism in the acamprosate treatment of certain sub-populations of alcohol dependent subjects.


Asunto(s)
Acamprosato/uso terapéutico , Disuasivos de Alcohol/uso terapéutico , Alcoholismo/tratamiento farmacológico , Modelos Animales de Enfermedad , Alcoholismo/genética , Alcoholismo/metabolismo , Animales , Tranportador Equilibrativo 1 de Nucleósido/genética , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Proteómica , Transducción de Señal , Resultado del Tratamiento
3.
Neuropharmacology ; 131: 58-67, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29225043

RESUMEN

Dysfunction of N-methyl-d-aspartate receptor (NMDAR) signaling in the nucleus accumbens (NAc) has been implicated in the pathophysiology of alcohol use disorders (AUD). Neurogranin (Ng), a calmodulin-binding protein, is exclusively expressed in the post-synapse, and mediates NMDAR driven synaptic plasticity by regulating the calcium-calmodulin (Ca2+-CaM) pathway. To study the functional role of Ng in AUD, we administrated behavior tests including Pavlovian instrument transfer (PIT), operant conditioning, and rotarod test using Ng null mice (Ng-/- mice). We used adeno-associated virus (AAV)-mediated Ng expression and pharmacological manipulation to validate behavioral responses in Ng-/- mice. The results from our multidisciplinary approaches demonstrated that deficit of Ng increases tolerance to NMDAR inhibition and elicit faster cue reactivity during PIT without changes in ethanol reward. Operant conditioning results demonstrated that Ng-/- mice self-administered significantly more ethanol and displayed reduced sensitivity to aversive motivation. We identified that ethanol exposure decreases mGluR5 (metabotropic glutamate receptor 5) expression in the NAc of Ng-/- mice and pharmacological inhibition of mGluR5 reverses NMDAR desensitization in Ng-/- mice. Together these findings specifically suggest that accumbal Ng plays an essential role in the counterbalance between NMDAR and mGluR5 signaling; which alters NMDAR resistance, and thereby altering aversive motivation for ethanol and may ultimately contribute to susceptibility for alcohol addiction.


Asunto(s)
Depresores del Sistema Nervioso Central/administración & dosificación , Comportamiento de Búsqueda de Drogas/fisiología , Etanol/administración & dosificación , Motivación/fisiología , Neurogranina/metabolismo , Núcleo Accumbens/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , 2-Amino-5-fosfonovalerato/análogos & derivados , 2-Amino-5-fosfonovalerato/farmacología , Animales , Condicionamiento Operante/efectos de los fármacos , Dependovirus/genética , Relación Dosis-Respuesta a Droga , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Motivación/efectos de los fármacos , Neurogranina/genética , Núcleo Accumbens/ultraestructura , Autoadministración , Sacarosa/administración & dosificación , Factores de Tiempo
4.
Sci Rep ; 7(1): 2496, 2017 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-28566752

RESUMEN

Precision medicine for alcohol use disorder (AUD) allows optimal treatment of the right patient with the right drug at the right time. Here, we generated multivariable models incorporating clinical information and serum metabolite levels to predict acamprosate treatment response. The sample of 120 patients was randomly split into a training set (n = 80) and test set (n = 40) five independent times. Treatment response was defined as complete abstinence (no alcohol consumption during 3 months of acamprosate treatment) while nonresponse was defined as any alcohol consumption during this period. In each of the five training sets, we built a predictive model using a least absolute shrinkage and section operator (LASSO) penalized selection method and then evaluated the predictive performance of each model in the corresponding test set. The models predicted acamprosate treatment response with a mean sensitivity and specificity in the test sets of 0.83 and 0.31, respectively, suggesting our model performed well at predicting responders, but not non-responders (i.e. many non-responders were predicted to respond). Studies with larger sample sizes and additional biomarkers will expand the clinical utility of predictive algorithms for pharmaceutical response in AUD.


Asunto(s)
Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Alcoholismo/tratamiento farmacológico , Biomarcadores/sangre , Metabolómica , Acamprosato/administración & dosificación , Adulto , Consumo de Bebidas Alcohólicas/sangre , Consumo de Bebidas Alcohólicas/epidemiología , Consumo de Bebidas Alcohólicas/patología , Alcoholismo/sangre , Alcoholismo/epidemiología , Alcoholismo/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Medicina de Precisión , Resultado del Tratamiento
5.
J Clin Psychopharmacol ; 36(6): 669-674, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27755217

RESUMEN

Although the precise drug mechanism of action of acamprosate remains unclear, its antidipsotropic effect is mediated in part through glutamatergic neurotransmission. We evaluated the effect of 4 weeks of acamprosate treatment in a cohort of 13 subjects with alcohol dependence (confirmed by a structured interview, Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision) on proton magnetic resonance spectroscopy glutamate levels in the midline anterior cingulate cortex (MACC). We compared levels of metabolites with a group of 16 healthy controls. The Pennsylvania Alcohol Craving Scale was used to assess craving intensity. At baseline, before treatment, the mean cerebrospinal fluid-corrected MACC glutamate (Glu) level was significantly elevated in subjects with alcohol dependence compared with controls (P = 0.004). Four weeks of acamprosate treatment reduced glutamate levels (P = 0.025), an effect that was not observed in subjects who did not take acamprosate. At baseline, there was a significant positive correlation between cravings, measured by the Pennsylvania Alcohol Craving Scale, and MACC (Glu) levels (P = 0.019). Overall, these data would suggest a normalizing effect of acamprosate on a hyperglutamatergic state observed in recently withdrawn patients with alcohol dependence and a positive association between MACC glutamate levels and craving intensity in early abstinence. Further research is needed to evaluate the use of these findings for clinical practice, including monitoring of craving intensity and individualized selection of treatment with antidipsotropic medications in subjects with alcohol dependence.


Asunto(s)
Disuasivos de Alcohol/farmacología , Alcoholismo/tratamiento farmacológico , Ácido Glutámico/metabolismo , Giro del Cíngulo/metabolismo , Taurina/análogos & derivados , Acamprosato , Adulto , Disuasivos de Alcohol/administración & dosificación , Alcoholismo/metabolismo , Femenino , Ácido Glutámico/efectos de los fármacos , Giro del Cíngulo/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Espectroscopía de Protones por Resonancia Magnética , Taurina/administración & dosificación , Taurina/farmacología , Resultado del Tratamiento , Adulto Joven
6.
J Neuroinflammation ; 13(1): 222, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27576583

RESUMEN

BACKGROUND: CD8 T cell-mediated blood-brain barrier (BBB) disruption is dependent on the effector molecule perforin. Human perforin has extensive single nucleotide variants (SNVs), the significance of which is not fully understood. These SNVs can result in reduced, but not ablated, perforin activity or expression. However, complete loss of perforin expression or activity results in the lethal disease familial hemophagocytic lymphohistiocytosis type 2 (FHL 2). In this study, we address the hypothesis that a single perforin allele can alter the severity of BBB disruption in vivo using a well-established model of CNS vascular permeability in C57Bl/6 mice. The results of this study provide insight into the significance of perforin SNVs in the human population. METHODS: We isolated the effect a single perforin allele has on CNS vascular permeability through the use of perforin-heterozygous (perforin+/-) C57BL/6 mice in the peptide-induced fatal syndrome (PIFS) model of immune-mediated BBB disruption. Seven days following Theiler's murine encephalomyelitis virus (TMEV) CNS infection, neuroinflammation and TMEV viral control were assessed through flow cytometric analysis and quantitative real-time PCR of the viral genome, respectively. Following immune-mediated BBB disruption, gadolinium-enhanced T1-weighted MRI, with 3D volumetric analysis, and confocal microscopy were used to define CNS vascular permeability. Finally, the open field behavior test was used to assess locomotor activity of mice following immune-mediated BBB disruption. RESULTS: Perforin-null mice had negligible CNS vascular permeability. Perforin-WT mice have extensive CNS vascular permeability. Interestingly, perforin-heterozygous mice had an intermediate level of CNS vascular permeability as measured by both gadolinium-enhanced T1-weighted MRI and fibrinogen leakage in the brain parenchyma. Differences in BBB disruption were not a result of increased CNS immune infiltrate. Additionally, TMEV was controlled in a perforin dose-dependent manner. Furthermore, a single perforin allele is sufficient to induce locomotor deficit during immune-mediated BBB disruption. CONCLUSIONS: Perforin modulates BBB disruption in a dose-dependent manner. This study demonstrates a potentially advantageous role for decreased perforin expression in reducing BBB disruption. This study also provides insight into the effect SNVs in a single perforin allele could have on functional deficit in neurological disease.


Asunto(s)
Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Dosificación de Gen/fisiología , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Animales , Barrera Hematoencefálica/virología , Encéfalo/virología , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Citotóxicas Formadoras de Poros/genética , Theilovirus/genética , Theilovirus/metabolismo
7.
Alcohol Clin Exp Res ; 40(8): 1609-16, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27439218

RESUMEN

BACKGROUND: Quantifying craving longitudinally during the course of withdrawal, early abstinence, and relapse is essential for optimal management of alcohol use disorder (AUD). In an effort to identify biological correlates of craving, we used proton magnetic resonance spectroscopy (1H-MRS) to investigate the correlation between craving and glutamate levels in the left dorsolateral prefrontal cortex (LDLPFC) of patients with AUD. METHODS: Participants underwent 1H-MRS of the LDLPFC with 2-dimensional J-resolved (2DJ) averaged PRESS. MRS data were processed with LCModel and cerebrospinal fluid (CSF)-corrected to generate metabolite concentrations. The Penn Alcohol Craving Scale (PACS) and the 30-day time line follow-back (TLFB 30) were used to quantify craving for alcohol and drinking patterns, respectively. RESULTS: There was a statistically significant positive correlation between CSF-corrected glutamate ([Glu]) levels and PACS scores (n = 14; p = 0.005). When PACS scores were dichotomized (< or ≥median = 16), [Glu] levels were significantly higher in the high- versus low-craving group (p = 0.007). In addition, there was a significant negative correlation between CSF-corrected N-acetyl aspartic acid ([NAA]) levels and mean number of drinks per drinking day in the past month (n = 13; TLFB 30; p = 0.012). When mean TLFB 30 was dichotomized (< or ≥median = 7.86), [NAA] levels were significantly lower in subjects that consumed more alcoholic beverages. There was no significant correlation between [Glu] and [NAA] levels with other measures of drinking behavior and or depression symptom severity. CONCLUSIONS: While limited by small sample size, these data suggest that glutamate levels in LDLPFC are associated with alcohol craving intensity in patients with AUD. Further study with larger sample size is needed to replicate this finding and evaluate the merits of glutamate spectroscopy as a biological correlate of alcohol craving intensity and a guide to treatment interventions.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Trastornos Relacionados con Alcohol/metabolismo , Ansia/fisiología , Ácido Glutámico/metabolismo , Corteza Prefrontal/metabolismo , Adulto , Abstinencia de Alcohol/tendencias , Trastornos Relacionados con Alcohol/diagnóstico por imagen , Trastornos Relacionados con Alcohol/terapia , Femenino , Humanos , Estudios Longitudinales , Espectroscopía de Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Corteza Prefrontal/diagnóstico por imagen , Centros de Tratamiento de Abuso de Sustancias/tendencias
8.
Alcohol Clin Exp Res ; 40(7): 1531-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27184383

RESUMEN

BACKGROUND: Major depression is one of the most prevalent psychiatry comorbidities of alcohol use disorders (AUD). As negative emotions can trigger craving and increase the risk of relapse, treatments that target both conditions simultaneously may augment treatment success. Previous studies showed a potential synergistic effect of Food and Drug Administration approved medication for AUD acamprosate and the antidepressant escitalopram. In this study, we investigated the effects of combining acamprosate and escitalopram on ethanol (EtOH) consumption in stress-induced depressed mice. METHODS: Forty singly housed C57BL/6J male mice were subjected to chronic unpredictable stress. In parallel, 40 group-housed male mice were subjected to normal husbandry. After 3 weeks, depressive- and anxiety-like behaviors and EtOH consumption were assessed. For the next 7 days, mice were injected with saline, acamprosate (200 mg/kg; twice/d), escitalopram (5 mg/kg; twice/d), or their combination (n = 9 to 11/drug group/stress group). Two-bottle choice limited-access drinking of 15% EtOH and tap water was performed 3 hours into dark phase immediately after the daily dark phase injection. EtOH drinking was monitored for another 7 days without drug administration. RESULTS: Mice subjected to the chronic unpredictable stress paradigm for 3 weeks showed apparent depression- and anxiety-like behaviors compared to their nonstressed counterparts including longer immobility time in the forced swim test and lower sucrose preference. Stressed mice also displayed higher EtOH consumption and preference in a 2-bottle choice drinking test. During the drug administration period, the escitalopram-only and combined drug groups showed significant reduction in EtOH consumption in nonstressed mice, while only the combined drug group showed significantly reduced consumption in stressed mice. However, such reduction did not persist into the postdrug administration period. CONCLUSIONS: The combination of acamprosate and escitalopram suppressed EtOH intake in both nonstressed and stressed mice; hence, this combination is potentially helpful for AUD individuals with or without comorbid depression to reduce alcohol use.


Asunto(s)
Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Citalopram/uso terapéutico , Taurina/análogos & derivados , Acamprosato , Animales , Ansiedad/complicaciones , Ansiedad/tratamiento farmacológico , Depresión/complicaciones , Depresión/tratamiento farmacológico , Quimioterapia Combinada , Masculino , Ratones , Estrés Psicológico/tratamiento farmacológico , Taurina/uso terapéutico
9.
Front Behav Neurosci ; 10: 46, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27014007

RESUMEN

The molecular mechanisms underlying the neuronal signaling changes in alcohol addiction and withdrawal are complex and multifaceted. The cortico-striatal circuit is highly implicated in these processes, and the striatum plays a significant role not only in the early stages of addiction, but in the developed-addictive state as well, including withdrawal symptoms. Transcriptional analysis is a useful method for determining changes in gene expression, however, the results do not always accurately correlate with protein levels. In this study, we employ label-free proteomic analysis to determine changes in protein expression within the striatum during chronic ethanol use and early withdrawal. The striatum, composed primarily of medium spiny GABAergic neurons, glutamatergic and dopaminergic nerve terminals and astrocytes, is relatively homogeneous for proteomic analysis. We were able to analyze more than 5000 proteins from both the dorsal (caudate and putamen) and ventral (nucleus accumbens) striatum and identified significant changes following chronic intermittent ethanol exposure and acute (8 h) withdrawal compared to ethanol naïve and ethanol exposure groups respectively. Our results showed significant changes in proteins involved in glutamate and opioid peptide signaling, and also uncovered novel pathways including mitochondrial function and lipid/cholesterol metabolism, as revealed by changes in electron transport chain proteins and RXR activation pathways. These results will be useful in the development of novel treatments for alcohol withdrawal and thereby aid in recovery from alcohol use disorder.

10.
Brain Behav ; 4(6): 903-14, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25365803

RESUMEN

BACKGROUND: Adenosine signaling has been implicated in several neurological and psychiatric disorders. Previously, we found that astrocytic excitatory amino acid transporter 2 (EAAT2) and aquaporin 4 (AQP4) are downregulated in the striatum of mice lacking type 1 equilibrative nucleoside transporter (ENT1). METHODS: To further investigate the gene expression profile in the striatum, we preformed Illumina Mouse Whole Genome BeadChip microarray analysis of the caudate-putamen (CPu) and nucleus accumbens (NAc) in ENT1 null mice. Gene expression was validated by RT-PCR, Western blot, and immunofluorescence. Using transgenic mice expressing enhanced green fluorescence protein (EGFP) under the control of the glial fibrillary acidic protein (GFAP) promoter, we examined EGFP expression in an ENT1 null background. RESULTS: Glial fibrillary acidic protein was identified as a top candidate gene that was reduced in ENT1 null mice compared to wild-type littermates. Furthermore, EGFP expression was significantly reduced in GFAP-EGFP transgenic mice in an ENT1 null background in both the CPu and NAc. Finally, pharmacological inhibition or siRNA knockdown of ENT1 in cultured astrocytes also reduced GFAP mRNA levels. CONCLUSIONS: Overall, our findings demonstrate that ENT1 regulates GFAP expression and possibly astrocyte function.


Asunto(s)
Astrocitos/metabolismo , Cuerpo Estriado/metabolismo , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Western Blotting , Línea Celular , Tranportador Equilibrativo 1 de Nucleósido/antagonistas & inhibidores , Tranportador Equilibrativo 1 de Nucleósido/genética , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Análisis por Micromatrices , Proteínas del Tejido Nervioso/genética , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Front Neurosci ; 8: 288, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25278825

RESUMEN

Peptides synthesized in endocrine cells in the gastrointestinal tract and neurons are traditionally considered regulators of metabolism, energy intake, and appetite. However, recent work has demonstrated that many of these peptides act on corticostriatal-limbic circuitry and, in turn, regulate addictive behaviors. Given that alcohol is a source of energy and an addictive substance, it is not surprising that increasing evidence supports a role for gut-brain peptides specifically in alcohol use disorders (AUD). In this review, we discuss the effects of several gut-brain peptides on alcohol-related behaviors and the potential mechanisms by which these gut-brain peptides may interfere with alcohol-induced changes in corticostriatal-limbic circuitry. This review provides a summary of current knowledge on gut-brain peptides focusing on five peptides: neurotensin, glucagon-like peptide 1, ghrelin, substance P, and neuropeptide Y. Our review will be helpful to develop novel therapeutic targets for AUD.

12.
Neuropharmacology ; 85: 482-92, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24929110

RESUMEN

Intracerebroventricular administration of neurotensin (NT) suppresses locomotor activity. However, the brain regions that mediate the locomotor depressant effect of NT and receptor subtype-specific mechanisms involved are unclear. Using a brain-penetrating, selective NT receptor type 1 (NTS1) agonist PD149163, we investigated the effect of systemic and brain region-specific NTS1 activation on locomotor activity. Systemic administration of PD149163 attenuated the locomotor activity of C57BL/6J mice both in a novel environment and in their homecage. However, mice developed tolerance to the hypolocomotor effect of PD149163 (0.1 mg/kg, i.p.). Since NTS1 is known to modulate dopaminergic signaling, we examined whether PD149163 blocks dopamine receptor-mediated hyperactivity. Pretreatment with PD149163 (0.1 or 0.05 mg/kg, i.p.) inhibited D2R agonist bromocriptine (8 mg/kg, i.p.)-mediated hyperactivity. D1R agonist SKF-81297 (8 mg/kg, i.p.)-induced hyperlocomotion was only inhibited by 0.1 mg/kg of PD149163. Since the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC) have been implicated in the behavioral effects of NT, we examined whether microinjection of PD149163 into these regions reduces locomotion. Microinjection of PD149163 (2 pmol) into the NAc, but not the mPFC suppressed locomotor activity. In summary, our results indicate that systemic and intra-NAc activation of NTS1 is sufficient to reduce locomotion and NTS1 activation inhibits D2R-mediated hyperactivity. Our study will be helpful to identify pharmacological factors and a possible therapeutic window for NTS1-targeted therapies for movement disorders.


Asunto(s)
Fármacos del Sistema Nervioso Central/farmacología , Actividad Motora/efectos de los fármacos , Neurotensina/análogos & derivados , Núcleo Accumbens/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Receptores de Neurotensina/agonistas , Animales , Benzazepinas/farmacología , Bromocriptina/farmacología , Agonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Ambiente , Vivienda para Animales , Masculino , Ratones Endogámicos C57BL , Microinyecciones , Actividad Motora/fisiología , Fuerza Muscular/efectos de los fármacos , Fuerza Muscular/fisiología , Neurotensina/farmacología , Núcleo Accumbens/fisiología , Corteza Prefrontal/fisiología , Distribución Aleatoria , Receptores Dopaminérgicos/metabolismo , Receptores de Neurotensina/metabolismo , Prueba de Desempeño de Rotación con Aceleración Constante
13.
Neuropsychopharmacology ; 39(10): 2432-40, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24755889

RESUMEN

Circadian rhythm and sleep disruptions occur frequently in individuals with alcohol use disorders (AUD) and present significant barriers to treatment. Recently, a variant of adenosine transporter, equilibrative nucleoside transporter 1 (ENT1), was associated with the co-occurrence of sleep problems and AUD. We have previously shown that mice lacking ENT1 (ENT1 KO) have reduced adenosine levels in the striatum and drink more alcohol compared with wild types (WT). However, it is unknown whether ENT1 deletion disrupts circadian rhythms, which may contribute to alcohol preference in ENT1 KO mice. Here we used these mice to determine whether endogenous adenosine regulates circadian genetic and behavioral rhythms and influences alcohol intake during chronodisruption. We examined circadian locomotor activity in ENT1 KO vs WT littermates and found that ENT1 KO mice were both active earlier and hyperactive compared with WT mice at night. We used real-time PCR and immunohistochemistry to estimate striatal clock gene levels and found that PER2 expression in the striatum was blunted by ENT1 deletion or A2A receptor (A2AR) antagonism. Next, we exposed ENT1 KO and WT mice to constant light (LL) and found further elevation in ethanol intake in ENT1 KO, but not in WT mice, supporting the notion that circadian dysfunction may contribute to increased alcohol intake in ENT1 KO mice. Finally, we showed that A2AR agonist administration normalized PER1 and PER2 expression and circadian locomotor activity in ENT1 KO mice. Together, our results demonstrate that adenosine signaling regulates cellular and behavioral circadian timing and influences alcohol intake during chronodisruption.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Ritmo Circadiano/fisiología , Cuerpo Estriado/fisiopatología , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Actividad Motora/fisiología , Receptor de Adenosina A2A/metabolismo , Agonistas del Receptor de Adenosina A2/farmacología , Antagonistas del Receptor de Adenosina A2/farmacología , Animales , Depresores del Sistema Nervioso Central/administración & dosificación , Ritmo Circadiano/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Tranportador Equilibrativo 1 de Nucleósido/genética , Etanol/administración & dosificación , Expresión Génica , Luz , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Proteínas Circadianas Period/metabolismo , Estimulación Luminosa
14.
PLoS One ; 9(2): e88818, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586402

RESUMEN

Adenosine is known to regulate bone production and resorption in humans and mice. Type 1 equilibrative nucleoside transporter (ENT1) is responsible for the majority of adenosine transport across the plasma membrane and is ubiquitously expressed in both humans and mice. However, the contribution of ENT1-mediated adenosine levels has not been studied in bone remodeling. With the recent identification of the importance of adenosine signaling in bone homeostasis, it is essential to understand the role of ENT1 to develop novel therapeutic compounds for bone disorders. Here we examined the effect of ENT1 deletion on bone density using X-ray, dual energy X-ray absorptiometry and micro-computerized tomography analysis. Our results show that bone density and bone mineral density is reduced in the lower thoracic and lumbar spine as well as the femur of old ENT1 null mice (>7 months) compared to wild-type littermates. Furthermore, we found increased mRNA expression of tartrate-resistant acid phosphatase (TRAP), an osteoclast marker, in isolated long bones from 10 month old ENT1 null mice compared to wild-type mice. In addition, aged ENT1 null mice displayed severe deficit in motor coordination and locomotor activity, which might be attributed to dysregulated bone density. Overall, our study suggests that ENT1-regulated adenosine signaling plays an essential role in lumbar spine and femur bone density.


Asunto(s)
Envejecimiento/fisiología , Densidad Ósea/genética , Densidad Ósea/fisiología , Tranportador Equilibrativo 1 de Nucleósido/genética , Eliminación de Gen , Envejecimiento/genética , Animales , Peso Corporal/genética , Peso Corporal/fisiología , Fémur/citología , Fémur/crecimiento & desarrollo , Fémur/fisiología , Regulación de la Expresión Génica , Vértebras Lumbares/citología , Vértebras Lumbares/crecimiento & desarrollo , Vértebras Lumbares/fisiología , Ratones , Actividad Motora/genética , Actividad Motora/fisiología , Tamaño de los Órganos/genética , Tamaño de los Órganos/fisiología , Osteoclastos/citología , Osteoclastos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Prueba de Desempeño de Rotación con Aceleración Constante
15.
J Neurochem ; 127(5): 691-700, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23745722

RESUMEN

Aging and the presence of cerebrovascular disease are associated with increased incidence of Alzheimer's disease. A common feature of aging and cerebrovascular disease is decreased endothelial nitric oxide (NO). We studied the effect of a loss of endothelium derived NO on amyloid precursor protein (APP) related phenotype in late middle aged (LMA) (14-15 month) endothelial nitric oxide synthase deficient (eNOS(-/-) ) mice. APP, ß-site APP cleaving enzyme (BACE) 1, and amyloid beta (Aß) levels were significantly higher in the brains of LMA eNOS(-/-) mice as compared with LMA wild-type controls. APP and Aß1-40 were increased in hippocampal tissue of eNOS(-/-) mice as compared with wild-type mice. LMA eNOS(-/-) mice displayed an increased inflammatory phenotype as compared with LMA wild-type mice. Importantly, LMA eNOS(-/-) mice performed worse in a radial arm maze test of spatial learning and memory as compared with LMA wild-type mice. These data suggest that chronic loss of endothelial NO may be an important contributor to both Aß related pathology and cognitive decline. Cardiovascular risk factors are associated with increased incidence of Alzheimer's disease (AD). A common feature of these risk factors is decreased endothelial nitric oxide (NO). We observed, in mice deficient in endothelial nitric oxide synthase, increased amyloid precursor protein (APP), ß-site APP cleaving enzyme 1, amyloid beta levels, microglial activation, and impaired spatial memory. This suggests chronic loss of endothelial NO may be an important contributor to the pathogenesis of sporadic AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Enfermedad de Alzheimer/epidemiología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encefalitis/epidemiología , Encefalitis/metabolismo , Encefalitis/patología , Hipocampo/irrigación sanguínea , Hipocampo/enzimología , Hipocampo/patología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Microvasos/enzimología , Microvasos/patología , Óxido Nítrico Sintasa de Tipo III/deficiencia , Fenotipo , Factores de Riesgo
16.
J Neurosci ; 33(10): 4329-38, 2013 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23467349

RESUMEN

Adenosine signaling has been implicated in the pathophysiology of many psychiatric disorders including alcoholism. Striatal adenosine A2A receptors (A2AR) play an essential role in both ethanol drinking and the shift from goal-directed action to habitual behavior. However, direct evidence for a role of striatal A2AR signaling in ethanol drinking and habit development has not been established. In the present study, we found that decreased A2AR-mediated CREB activity in the dorsomedial striatum (DMS) enhanced initial behavioral acquisition of goal-directed behaviors and the vulnerability to progress to excessive ethanol drinking during operant conditioning in mice lacking ethanol-sensitive adenosine transporter ENT1 (ENT1(-/-)). Using mice expressing ß-galactosidase (lacZ) under the control of seven repeated CRE sites in both genotypes (CRE-lacZ/ENT1(+/+) mice and CRE-lacZ/ENT1(-/-) mice) and the dominant-negative form of CREB, we found that reduced CREB activity in the DMS was causally associated with decreased A2AR signaling and increased goal-directed ethanol drinking. Finally, we have demonstrated that the A2AR antagonist ZM241385 dampened protein kinase A activity-mediated signaling in the DMS and promoted excessive ethanol drinking in ENT1(+/+) mice, but not in ENT1(-/-) mice. Our results indicate that A2AR-mediated CREB signaling in the DMS is a key determinant in enhancing the development of goal-directed ethanol drinking in mice.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Conducta de Elección/fisiología , Cuerpo Estriado/fisiología , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Objetivos , Receptor de Adenosina A2A/metabolismo , Consumo de Bebidas Alcohólicas/genética , Análisis de Varianza , Animales , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Conducta de Elección/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Cuerpo Estriado/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Tranportador Equilibrativo 1 de Nucleósido/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esquema de Refuerzo , Transducción de Señal/efectos de los fármacos , Sacarosa/administración & dosificación , Edulcorantes/administración & dosificación , Triazinas/farmacología , Triazoles/farmacología , beta-Galactosidasa/metabolismo
17.
Behav Brain Res ; 244: 58-65, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23376703

RESUMEN

An herb derived compound, levo-tetrahydropalmatine (L-THP), attenuates self-administration of cocaine and opiates in rodents. Since L-THP mainly antagonizes dopamine D2 receptors (D2R) in the brain, it is likely to regulate other addictive behaviors as well. Here, we examined whether L-THP regulates ethanol drinking in C57BL/6J mice using a two-bottle choice drinking experiment. L-THP treated mice consumed less ethanol compared to vehicle-treated mice during the 15% ethanol drinking session while water consumption remained similar between each group. We then examined the molecular basis underlying the pharmacological effect of L-THP in mice. Our results indicated that a single injection of L-THP increased active phosphorylated forms of PKA, AKT and ERK in the caudate-putamen (CPu), but not in the nucleus accumbens (NAc), of alcohol naïve mice. Interestingly, we found that systematic treatment with L-THP for 4 consecutive days while mice were drinking 15% ethanol increased pPKA levels in the CPu, but not in the NAc. In contrast to the effect of acute L-THP treatment, no differences were detected for pAKT or pERK in either striatal regions. Together, our findings suggest that reduction of ethanol drinking by L-THP treatment is possibly correlated with D2R-mediated PKA signaling in the CPu.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Alcaloides de Berberina/farmacología , Cuerpo Estriado/efectos de los fármacos , Antagonistas de Dopamina/farmacología , Antagonistas de los Receptores de Dopamina D2 , Transducción de Señal/efectos de los fármacos , Consumo de Bebidas Alcohólicas/sangre , Consumo de Bebidas Alcohólicas/psicología , Animales , Conducta de Elección/efectos de los fármacos , Conducta de Elección/fisiología , Cuerpo Estriado/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Ratones , Proteína Oncogénica v-akt/metabolismo
18.
Neuropsychopharmacology ; 38(3): 437-45, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23032072

RESUMEN

Adenosine signaling is implicated in several neuropsychiatric disorders, including alcoholism. Among its diverse functions in the brain, adenosine regulates glutamate release and has an essential role in ethanol sensitivity and preference. However, the molecular mechanisms underlying adenosine-mediated glutamate signaling in neuroglial interaction remain elusive. We have previously shown that mice lacking the ethanol-sensitive adenosine transporter, type 1 equilibrative nucleoside transporter (ENT1), drink more ethanol compared with wild-type mice and have elevated striatal glutamate levels. In addition, ENT1 inhibition or knockdown reduces glutamate transporter expression in cultured astrocytes. Here, we examined how adenosine signaling in astrocytes contributes to ethanol drinking. Inhibition or deletion of ENT1 reduced the expression of type 2 excitatory amino-acid transporter (EAAT2) and the astrocyte-specific water channel, aquaporin 4 (AQP4). EAAT2 and AQP4 colocalization was also reduced in the striatum of ENT1 null mice. Ceftriaxone, an antibiotic compound known to increase EAAT2 expression and function, elevated not only EAAT2 but also AQP4 expression in the striatum. Furthermore, ceftriaxone reduced ethanol drinking, suggesting that ENT1-mediated downregulation of EAAT2 and AQP4 expression contributes to excessive ethanol consumption in our mouse model. Overall, our findings indicate that adenosine signaling regulates EAAT2 and astrocytic AQP4 expressions, which control ethanol drinking in mice.


Asunto(s)
Adenosina/fisiología , Consumo de Bebidas Alcohólicas/metabolismo , Acuaporina 4/biosíntesis , Astrocitos/fisiología , Cuerpo Estriado/metabolismo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/fisiopatología , Animales , Acuaporina 4/antagonistas & inhibidores , Astrocitos/patología , Línea Celular , Cuerpo Estriado/fisiopatología , Regulación hacia Abajo/genética , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética
19.
Bioanalysis ; 4(14): 1787-804, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22877223

RESUMEN

The ongoing development of animal models of neurological and psychiatric disorders in combination with the development of advanced nuclear magnetic resonance (NMR) techniques and instrumentation has led to increased use of in vivo proton NMR spectroscopy ((1)H-MRS) for neurochemical analyses. (1)H-MRS is one of only a few analytical methods that can assay in vivo and longitudinal neurochemical changes associated with neurological and psychiatric diseases, with the added advantage of being a technique that can be utilized in both preclinical and clinical studies. In this review, recent progress in the use of (1)H-MRS to investigate animal models of neurological and psychiatric disorders is summarized with examples from the literature and our own work.


Asunto(s)
Biomarcadores/análisis , Química Encefálica , Trastornos Mentales/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Resonancia Magnética Nuclear Biomolecular/métodos , Alcoholismo/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Humanos , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Noqueados , Trastornos del Humor/metabolismo , Esclerosis Múltiple/metabolismo , Ratas
20.
Neuropharmacology ; 62(8): 2480-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22616110

RESUMEN

Acamprosate is clinically used to treat alcohol-dependent patients. While the molecular and pharmacological mechanisms of acamprosate remain unclear, it has been shown to regulate γ-aminobutyric acid (GABA) or glutamate levels in the cortex and striatum. To investigate the effect of acamprosate on brain metabolites in the medial prefrontal cortex (mPFC) and nucleus accumbens (NAc), we employed in vivo 16.4 T proton magnetic resonance spectroscopy. We utilized type 1 equilibrative nucleoside transporter (ENT1) null mice since acamprosate attenuates ethanol drinking in these mice. Our findings demonstrated that ethanol withdrawal reduced GABA levels and increased phosphorylated choline compounds in the mPFC of both wild-type and ENT1 null mice. Notably, acamprosate normalized these withdrawal-induced changes only in ENT1 null mice. In the NAc, ethanol withdrawal increased glutamate and glutamine (Glx) levels only in wild-type mice. Interestingly, acamprosate reduced Glx levels in the NAc compared to the withdrawal state in both genotypes. These results provide a molecular basis for the pharmacological effect of acamprosate in the cortical-striatal circuit.


Asunto(s)
Encéfalo/efectos de los fármacos , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Etanol/efectos adversos , Ácido Glutámico/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Taurina/análogos & derivados , Ácido gamma-Aminobutírico/metabolismo , Acamprosato , Animales , Encéfalo/metabolismo , Tranportador Equilibrativo 1 de Nucleósido/genética , Espectroscopía de Resonancia Magnética , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Taurina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...