Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Neurobiol Learn Mem ; 118: 162-6, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25543025

RESUMEN

Increased training often results in stronger memories but the neural changes responsible for these stronger memories are poorly understood. It is proposed here that higher levels of training that result in stronger memories recruit additional cell signaling cascades. This study specifically examined if c-Jun N-terminal kinase 1 (JNK1) is involved in the formation of stronger fear conditioning memories. Wildtype (WT), JNK1 heterozygous (Het), and JNK1 knockout (KO) mice were fear conditioned with 1 trial, 2 trials, or 4 trials. All mice learned both contextual (hippocampus-dependent) and cued (hippocampus-independent) fear conditioning but for contextual fear conditioning only, the JNK1 KO mice did not show higher levels of learning with increased trials. That is, WT mice showed a significant linear increase in contextual fear conditioning as training trials increased from 1 to 2 to 4 trials whereas KO mice showed the same level of contextual fear conditioning as WT mice for 1 trial training but did not have increased levels of contextual fear conditioning with additional trials. These data suggest that JNK1 may not be critical for learning but when higher levels of hippocampus-dependent learning occur, JNK1 signaling is recruited and is necessary for stronger hippocampus-dependent memory formation.


Asunto(s)
Condicionamiento Clásico/fisiología , Miedo/fisiología , Aprendizaje/fisiología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/fisiología , Transducción de Señal , Animales , Femenino , Hipocampo/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/genética , Actividad Motora
2.
Nicotine Tob Res ; 17(6): 690-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25358661

RESUMEN

INTRODUCTION: Cigarette smoking alters a variety of endocrine systems including thyroid hormones. Altered thyroid hormone signaling may lead to a subclinical or overt hypothyroid condition that could contribute to nicotine withdrawal-related symptoms, such as cognitive deficits. Thus, normalizing thyroid hormone levels may represent a novel therapeutic target for ameliorating nicotine withdrawal-associated cognitive deficits. METHODS: The current studies conducted an analysis of serum thyroid hormone levels after chronic and withdrawal from chronic nicotine treatment in C57BL/6J mice using an enzyme-linked immunosorbent assay. The present studies also evaluated the effect of synthetic thyroid hormone (levothyroxine) on contextual and cued memory. RESULTS: The current studies found that nicotine withdrawal reduces secreted thyroid hormone levels by 9% in C57BL/6J mice. Further, supplemental thyroid hormone not only enhanced memory in naïve animals, but also ameliorated deficits in hippocampus-dependent learning associated with nicotine withdrawal. CONCLUSIONS: These results suggest that smokers attempting to quit should be monitored closely for changes in thyroid function. If successfully treated, normalization of thyroid hormone levels may ameliorate some deficits associated with nicotine withdrawal and this may lead to higher rates of successful abstinence.


Asunto(s)
Condicionamiento Psicológico/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipotiroidismo/sangre , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Síndrome de Abstinencia a Sustancias/sangre , Glándula Tiroides/efectos de los fármacos , Tiroxina/farmacología , Animales , Miedo/efectos de los fármacos , Hipotiroidismo/psicología , Aprendizaje/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Nicotina/efectos adversos , Agonistas Nicotínicos/efectos adversos , Síndrome de Abstinencia a Sustancias/etiología , Síndrome de Abstinencia a Sustancias/psicología , Hormonas Tiroideas/sangre , Hormonas Tiroideas/farmacología , Tiroxina/sangre , Tabaquismo
3.
J Neurosci ; 33(18): 7728-41, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23637165

RESUMEN

Baseline and nicotine-modulated behaviors were assessed in mice harboring a null mutant allele of the nicotinic acetylcholine receptor (nAChR) subunit gene α2 (Chrna2). Homozygous Chrna2(-/-) mice are viable, show expected sex and Mendelian genotype ratios, and exhibit no gross neuroanatomical abnormalities. A broad range of behavioral tests designed to assess genotype-dependent effects on anxiety (elevated plus maze and light/dark box), motor coordination (narrow bean traverse and gait), and locomotor activity revealed no significant differences between mutant mice and age-matched wild-type littermates. Furthermore, a panel of tests measuring traits, such as body position, spontaneous activity, respiration, tremors, body tone, and startle response, revealed normal responses for Chrna2-null mutant mice. However, Chrna2(-/-) mice do exhibit a mild motor or coordination phenotype (a decreased latency to fall during the accelerating rotarod test) and possess an increased sensitivity to nicotine-induced analgesia in the hotplate assay. Relative to wild-type, Chrna2(-/-) mice show potentiated nicotine self-administration and withdrawal behaviors and exhibit a sex-dependent enhancement of nicotine-facilitated cued, but not trace or contextual, fear conditioning. Overall, our results suggest that loss of the mouse nAChR α2 subunit has very limited effects on baseline behavior but does lead to the potentiation of several nicotine-modulated behaviors.


Asunto(s)
Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Receptores Nicotínicos/deficiencia , Síndrome de Abstinencia a Sustancias/fisiopatología , Análisis de Varianza , Animales , Ansiedad/fisiopatología , Condicionamiento Clásico/efectos de los fármacos , Esquema de Medicación , Reacción de Fuga/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Miedo/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/efectos adversos , Neurotransmisores/metabolismo , Desempeño Psicomotor/efectos de los fármacos , Tiempo de Reacción , Reflejo/efectos de los fármacos , Reflejo de Sobresalto/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/etiología
4.
Neurobiol Learn Mem ; 107: 108-32, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23973448

RESUMEN

Addiction is a chronic disorder marked by long-lasting maladaptive changes in behavior and in reward system function. However, the factors that contribute to the behavioral and biological changes that occur with addiction are complex and go beyond reward. Addiction involves changes in cognitive control and the development of disruptive drug-stimuli associations that can drive behavior. A reason for the strong influence drugs of abuse can exert on cognition may be the striking overlap between the neurobiological substrates of addiction and of learning and memory, especially areas involved in declarative memory. Declarative memories are critically involved in the formation of autobiographical memories, and the ability of drugs of abuse to alter these memories could be particularly detrimental. A key structure in this memory system is the hippocampus, which is critically involved in binding multimodal stimuli together to form complex long-term memories. While all drugs of abuse can alter hippocampal function, this review focuses on nicotine. Addiction to tobacco products is insidious, with the majority of smokers wanting to quit; yet the majority of those that attempt to quit fail. Nicotine addiction is associated with the presence of drug-context and drug-cue associations that trigger drug seeking behavior and altered cognition during periods of abstinence, which contributes to relapse. This suggests that understanding the effects of nicotine on learning and memory will advance understanding and potentially facilitate treating nicotine addiction. The following sections examine: (1) how the effects of nicotine on hippocampus-dependent learning change as nicotine administration transitions from acute to chronic and then to withdrawal from chronic treatment and the potential impact of these changes on addiction, (2) how nicotine usurps the cellular mechanisms of synaptic plasticity, (3) the physiological changes in the hippocampus that may contribute to nicotine withdrawal deficits in learning, and (4) the role of genetics and developmental stage (i.e., adolescence) in these effects.


Asunto(s)
Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Animales , Cognición/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Humanos , Aprendizaje/fisiología , Memoria/fisiología , Plasticidad Neuronal/efectos de los fármacos , Tabaquismo/fisiopatología
5.
Neurobiol Learn Mem ; 109: 151-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24457151

RESUMEN

Acute nicotine enhances hippocampus-dependent learning through nicotine binding to ß2-containing nicotinic acetylcholine receptors (nAChRs), but it is unclear if nicotine is targeting processes involved in short-term memory (STM) leading to a strong long-term memory (LTM) or directly targeting LTM. In addition, the molecular mechanisms involved in the effects of nicotine on learning are unknown. Previous research indicates that protein kinase A (PKA), extracellular signal-regulated kinase 1/2 (ERK1/2), and protein synthesis are crucial for LTM. Therefore, the present study examined the effects of nicotine on STM and LTM and the involvement of PKA, ERK1/2, and protein synthesis in the nicotine-induced enhancement of hippocampus-dependent contextual learning in C57BL/6J mice. The protein synthesis inhibitor anisomycin impaired contextual conditioning assessed at 4 h but not 2 h post-training, delineating time points for STM (2 h) and LTM (4 h and beyond). Nicotine enhanced contextual conditioning at 4, 8, and 24 h but not 2 h post-training, indicating nicotine specifically enhances LTM but not STM. Furthermore, nicotine did not rescue deficits in contextual conditioning produced by anisomycin, suggesting that the nicotine enhancement of contextual conditioning occurs through a protein synthesis-dependent mechanism. In addition, inhibition of dorsal hippocampal PKA activity blocked the effect of acute nicotine on learning, and nicotine shifted the timing of learning-related PKA and ERK1/2 activity in the dorsal and ventral hippocampus. Thus, the present results suggest that nicotine specifically enhances LTM through altering the timing of PKA and ERK1/2 signaling in the hippocampus, and suggests that the timing of PKA and ERK1/2 activity could contribute to the strength of memories.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/efectos de los fármacos , Memoria a Largo Plazo/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Animales , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Hipocampo/enzimología , Hipocampo/fisiología , Masculino , Memoria a Largo Plazo/fisiología , Memoria a Corto Plazo/efectos de los fármacos , Memoria a Corto Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo
6.
Learn Mem ; 19(8): 319-24, 2012 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-22802593

RESUMEN

Growth arrest and DNA damage-inducible ß (Gadd45b) has been shown to be involved in DNA demethylation and may be important for cognitive processes. Gadd45b is abnormally expressed in subjects with autism and psychosis, two disorders associated with cognitive deficits. Furthermore, several high-throughput screens have identified Gadd45b as a candidate plasticity-related gene. However, a direct demonstration of a link between Gadd45b and memory has not been established. The current studies first determined whether expression of the Gadd45 family of genes was affected by contextual fear conditioning. Gadd45b, and to a lesser extent Gadd45g, were up-regulated in the hippocampus following contextual fear conditioning, whereas Gadd45a was not. Next, Gadd45b knockout mice were tested for contextual and cued fear conditioning. Gadd45b knockout mice exhibited a significant deficit in long-term contextual fear conditioning; however, they displayed normal levels of short-term contextual fear conditioning. No differences between Gadd45b knockout and wild-type mice were observed in cued fear conditioning. Because cued fear conditioning is hippocampus independent, while contextual fear conditioning is hippocampus dependent, the current studies suggest that Gadd45b may be important for long-term hippocampus-dependent memory storage. Therefore, Gadd45b may be a novel therapeutic target for the cognitive deficits associated with many neurodevelopmental, neurological, and psychiatric disorders.


Asunto(s)
Antígenos de Diferenciación/genética , Hipocampo/fisiología , Trastornos de la Memoria , Memoria a Largo Plazo/fisiología , Análisis de Varianza , Animales , Antígenos de Diferenciación/metabolismo , Condicionamiento Psicológico/fisiología , Electrochoque/efectos adversos , Miedo/fisiología , Regulación de la Expresión Génica/genética , Masculino , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , ARN Mensajero/metabolismo
7.
PLoS One ; 14(8): e0220125, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31408457

RESUMEN

BIN1 is the most important risk locus for Late Onset Alzheimer's Disease (LOAD), after ApoE. BIN1 AD-associated SNPs correlate with Tau deposition as well as with brain atrophy. Furthermore, the level of neuronal-specific BIN1 isoform 1 protein is decreased in sporadic AD cases in parallel with neuronal loss, despite an overall increase in BIN1 total mRNA. To address the relationship between reduction of BIN1 and neuronal cell loss in the context of Tau pathology, we knocked-down endogenous murine Bin1 via stereotaxic injection of AAV-Bin1 shRNA in the hippocampus of mice expressing Tau P301S (PS19). We observed a statistically significant reduction in the number of neurons in the hippocampus of mice injected with AAV-Bin1 shRNA in comparison with mice injected with AAV control. To investigate whether neuronal loss is due to deletion of Bin1 selectively in neurons in presence Tau P301S, we bred Bin1flox/flox with Thy1-Cre and subsequently with PS19 mice. Mice lacking neuronal Bin1 and expressing Tau P301S showed increased mortality, without increased neuropathology, when compared to neuronal Bin1 and Tau P301S-expressing mice. The loss of Bin1 isoform 1 resulted in reduced excitability in primary neurons in vitro, reduced neuronal c-fos expression as well as in altered microglia transcriptome in vivo. Taken together, our data suggest that the contribution of genetic variation in BIN1 locus to AD risk could result from a cell-autonomous reduction of neuronal excitability due to Bin1 decrease, exacerbated by the presence of aggregated Tau, coupled with a non-cell autonomous microglia activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Enfermedad de Alzheimer/patología , Biomarcadores/metabolismo , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Proteínas del Tejido Nervioso/fisiología , Neuronas/patología , Proteínas Supresoras de Tumor/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Conducta Animal , Encéfalo/metabolismo , Femenino , Perfilación de la Expresión Génica , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Ratas , Proteínas tau/metabolismo
8.
Neurosci Lett ; 627: 61-4, 2016 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-27235579

RESUMEN

Acute nicotine enhances hippocampus-dependent learning. Identifying how acute nicotine improves learning will aid in understanding how nicotine facilitates the development of maladaptive memories that contribute to drug-seeking behaviors, help development of medications to treat disorders associated with cognitive decline, and advance understanding of the neurobiology of learning and memory. The effects of nicotine on learning may involve recruitment of signaling through the c-Jun N-terminal kinase family (JNK 1-3). Learning in the presence of acute nicotine increases the transcription of mitogen-activated protein kinase 8 (MAPK8, also known as JNK1), likely through a CREB-dependent mechanism. The functional significance of JNK1 in the effects of acute nicotine on learning, however, is unknown. The current studies undertook a backward genetic approach to determine the functional contribution JNK1 protein makes to nicotine-enhanced contextual fear conditioning. JNK1 wildtype (WT) and knockout (KO) mice were administered acute nicotine prior to contextual and cued fear conditioning. 24h later, mice were evaluated for hippocampus-dependent (contextual fear conditioning) and hippocampus-independent (cued fear conditioning) memory. Nicotine selectively enhanced contextual conditioning in WT mice, but not in KO mice. Nicotine had no effect on hippocampus-independent learning in either genotype. JNK1 KO and WT mice given saline showed similar levels of learning. These data suggest that JNK1 may be recruited by nicotine and is functionally necessary for the acute effects of nicotine on learning and memory.


Asunto(s)
Miedo/efectos de los fármacos , Miedo/fisiología , Proteína Quinasa 8 Activada por Mitógenos/fisiología , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Animales , Condicionamiento Clásico , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/genética
9.
Curr Top Behav Neurosci ; 28: 1-52, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27305922

RESUMEN

Animal models provide preclinical tools to investigate the causal role of genetic mutations and environmental factors in the etiology of autism spectrum disorder (ASD). Knockout and humanized knock-in mice, and more recently knockout rats, have been generated for many of the de novo single gene mutations and copy number variants (CNVs) detected in ASD and comorbid neurodevelopmental disorders. Mouse models incorporating genetic and environmental manipulations have been employed for preclinical testing of hypothesis-driven pharmacological targets, to begin to develop treatments for the diagnostic and associated symptoms of autism. In this review, we summarize rodent behavioral assays relevant to the core features of autism, preclinical and clinical evaluations of pharmacological interventions, and strategies to improve the translational value of rodent models of autism.


Asunto(s)
Trastorno Autístico/tratamiento farmacológico , Trastorno Autístico/genética , Modelos Animales de Enfermedad , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Ratones , Ratones Noqueados , Fenotipo , Conducta Social , Investigación Biomédica Traslacional
10.
eNeuro ; 3(1)2016.
Artículo en Inglés | MEDLINE | ID: mdl-27022628

RESUMEN

Fragile X syndrome is a neurodevelopmental disorder characterized by mild-to-severe cognitive deficits. The complete absence of Fmr1 and its protein product in the mouse model of fragile X (Fmr1 KO) provides construct validity. A major conundrum in the field is the remarkably normal performance of Fmr1 mice on cognitive tests in most reports. One explanation may be insufficiently challenging cognitive testing procedures. Here we developed a delayed nonmatching to position touchscreen task to test the hypothesis that paradigms placing demands on working memory would reveal robust and replicable cognitive deficits in the Fmr1 KO mouse. We first tested Fmr1 KO mice (Fmr1) and their wild-type (WT) littermates in a simple visual discrimination task, followed by assessment of reversal learning. We then tested Fmr1 and WT mice in a new touchscreen nonmatch to position task and subsequently challenged their working memory abilities by adding delays, representing a higher cognitive load. The performance by Fmr1 KO mice was equal to WTs on both touchscreen tasks. Last, we replicated previous reports of normal performance by Fmr1 mice on Morris water maze spatial navigation and reversal. These results indicate that, while the Fmr1 mouse model effectively recapitulates many molecular and cellular aspects of fragile X syndrome, the cognitive profile of Fmr1 mice generally does not recapitulate the primary cognitive deficits in the human syndrome, even when diverse and challenging tasks are imposed.


Asunto(s)
Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/fisiología , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/psicología , Memoria a Corto Plazo/fisiología , Animales , Conducta de Elección/fisiología , Discriminación en Psicología/fisiología , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Aprendizaje Inverso/fisiología
11.
J Med Chem ; 59(7): 3231-48, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-26937601

RESUMEN

We describe successful efforts to optimize the in vivo profile and address off-target liabilities of a series of BACE1 inhibitors represented by 6 that embodies the recently validated fused pyrrolidine iminopyrimidinone scaffold. Employing structure-based design, truncation of the cyanophenyl group of 6 that binds in the S3 pocket of BACE1 followed by modification of the thienyl group in S1 was pursued. Optimization of the pyrimidine substituent that binds in the S2'-S2″ pocket of BACE1 remediated time-dependent CYP3A4 inhibition of earlier analogues in this series and imparted high BACE1 affinity. These efforts resulted in the discovery of difluorophenyl analogue 9 (MBi-4), which robustly lowered CSF and cortex Aß40 in both rats and cynomolgus monkeys following a single oral dose. Compound 9 represents a unique molecular shape among BACE inhibitors reported to potently lower central Aß in nonrodent preclinical species.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Diseño de Fármacos , Compuestos Heterocíclicos/química , Iminas/química , Péptidos beta-Amiloides/líquido cefalorraquídeo , Animales , Corteza Cerebral/metabolismo , Inhibidores Enzimáticos/farmacología , Macaca fascicularis , Estructura Molecular , Ratas , Relación Estructura-Actividad
12.
Neurosci Biobehav Rev ; 57: 252-63, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26344666

RESUMEN

Cigarette smoking is common despite its adverse effects on health, such as cardiovascular disease and stroke. Understanding the mechanisms that contribute to the addictive properties of nicotine makes it possible to target them to prevent the initiation of smoking behavior and/or increase the chance of successful quit attempts. While highly addictive, nicotine is not generally considered to be as reinforcing as other drugs of abuse. There are likely other mechanisms at work that contribute to the addictive liability of nicotine. Nicotine modulates aspects of the endocrine system, including the thyroid, which is critical for normal cognitive functioning. It is possible that nicotine's effects on thyroid function may alter learning and memory, and this may underlie some of its addictive potential. Here, we review the literature on thyroid function and cognition, with a focus on how nicotine alters thyroid hormone signaling and the potential impact on cognition. Changes in cognition are a major symptom of nicotine addiction. Current anti-smoking therapies have modest success at best. If some of the cognitive effects of nicotine are mediated through the thyroid hormone system, then thyroid hormone agonists may be novel treatments for smoking cessation therapies. The content of this review is important because it clarifies the relationship between smoking and thyroid function, which has been ill-defined in the past. This review is timely because the reduction in smoking rates we have seen in recent decades, due to public awareness campaigns and public smoking bans, has leveled off in recent years. Therefore, novel treatment approaches are needed to help reduce smoking rates further.


Asunto(s)
Cognición/efectos de los fármacos , Nicotina/farmacología , Transducción de Señal/efectos de los fármacos , Hormonas Tiroideas/metabolismo , Tabaquismo/metabolismo , Humanos
13.
Neuropharmacology ; 93: 155-63, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25666034

RESUMEN

Cigarette smoking is common despite adverse health effects. Nicotine's effects on learning may contribute to addiction by enhancing drug-context associations. Effects of nicotine on learning could be direct or could occur by altering systems that modulate cognition. Because thyroid signaling can alter cognition and nicotine/smoking may change thyroid function, nicotine could affect learning through changes in thyroid signaling. These studies investigate the functional contributions of thyroid receptor (TR) subtypes ß and α1 to nicotine-enhanced learning and characterize the effects of acute nicotine and learning on thyroid hormone levels. We conducted a high throughput screen of transcription factor activity to identify novel targets that may contribute to the effects of nicotine on learning. Based on these results, which showed that combined nicotine and learning uniquely acted to increase TR activation, we identified TRs as potential targets of nicotine. Further analyses were conducted to determine the individual and combined effects of nicotine and learning on thyroid hormone levels, but no changes were seen. Next, to determine the role of TRß and TRα1 in the effects of nicotine on learning, mice lacking the TRß or TRα1 gene and wildtype littermates were administered acute nicotine prior to fear conditioning. Nicotine enhanced contextual fear conditioning in TRα1 knockout mice and wildtypes from both lines but TRß knockout mice did not show nicotine-enhanced learning. This finding supports involvement of TRß signaling in the effect of acute nicotine on hippocampus-dependent memory. Acute nicotine enhances learning and these effects may involve processes regulated by the transcription factor TRß.


Asunto(s)
Hipocampo/efectos de los fármacos , Memoria/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Receptores beta de Hormona Tiroidea/metabolismo , Estimulación Acústica , Animales , Reacción de Prevención/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Miedo/efectos de los fármacos , Femenino , Hipocampo/fisiología , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Análisis por Matrices de Proteínas , Reflejo de Sobresalto/efectos de los fármacos , Receptores alfa de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/genética , Hormonas Tiroideas/sangre , Factores de Transcripción/metabolismo
14.
Intractable Rare Dis Res ; 3(4): 118-33, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25606362

RESUMEN

Fragile X Syndrome (FXS) is a commonly inherited form of intellectual disability and one of the leading genetic causes for autism spectrum disorder. Clinical symptoms of FXS can include impaired cognition, anxiety, hyperactivity, social phobia, and repetitive behaviors. FXS is caused by a CGG repeat mutation which expands a region on the X chromosome containing the FMR1 gene. In FXS, a full mutation (> 200 repeats) leads to hypermethylation of FMR1, an epigenetic mechanism that effectively silences FMR1 gene expression and reduces levels of the FMR1 gene product, fragile X mental retardation protein (FMRP). FMRP is an RNA-binding protein that is important for the regulation of protein expression. In an effort to further understand how loss of FMR1 and FMRP contribute to FXS symptomology, several FXS animal models have been created. The most well characterized rodent model is the Fmr1 knockout (KO) mouse, which lacks FMRP protein due to a disruption in its Fmr1 gene. Here, we review the behavioral phenotyping of the Fmr1 KO mouse to date, and discuss the clinical relevance of this mouse model to the human FXS condition. While much remains to be learned about FXS, the Fmr1 KO mouse is a valuable tool for understanding the repercussions of functional loss of FMRP and assessing the efficacy of pharmacological compounds in ameliorating the molecular and behavioral phenotypes relevant to FXS.

15.
Behav Neurosci ; 127(2): 303-10, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23565938

RESUMEN

Nicotine is a widely used addictive drug, with an estimated 73 million Americans 12 years of age or older having used a tobacco product in the last month, despite documented risks to personal health. Nicotine alters cognitive processes, which include effects on attention and impulsivity, a mechanism that may contribute to the addictive properties of the drug. Individuals with a variety of psychological disorders ranging from attention deficit hyperactivity disorder (ADHD) to schizophrenia smoke at a higher rate than the rest of the population and show deficits in impulse control. The present studies evaluated the effects of acute, chronic, and withdrawal from chronic nicotine on an operant task that measured premature and signaled nose pokes, as well as performance efficiency in C57BL/6J mice. Results indicate that acute nicotine (0.09 mg/kg intraperitoneally) does not alter the acquisition of the task, but does significantly increase performance efficiency once the behavior has been learned. In contrast, chronic nicotine (0, 6.3, 12.6, and 36 mg/kg/day subcutaneously) and withdrawal from chronic nicotine had no effect on performance efficiency. These results suggest that initial nicotine use may have beneficial effects on inhibitory control, but these effects are not maintained with chronic nicotine consumption as tolerance develops. The findings may provide an explanation for higher rates of smoking in patients with impulse control issues, as the smoking may represent an initial attempt at self-medication.


Asunto(s)
Conducta Animal/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Nicotina/efectos adversos , Síndrome de Abstinencia a Sustancias/psicología , Animales , Atención/efectos de los fármacos , Condicionamiento Operante/fisiología , Señales (Psicología) , Masculino , Ratones , Nicotina/administración & dosificación , Tiempo de Reacción/efectos de los fármacos
16.
Int J Alzheimers Dis ; 2013: 823528, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23573456

RESUMEN

Substantial evidence implicates ß-amyloid (Aß) peptides in the etiology of Alzheimer's disease (AD). Aß is produced by the proteolytic cleavage of the amyloid precursor protein by ß- and γ-secretase suggesting that γ-secretase inhibition may provide therapeutic benefit for AD. Although many γ-secretase inhibitors have been shown to be potent at lowering Aß, some have also been shown to have side effects following repeated administration. All of these side effects can be attributed to altered Notch signaling, another γ-secretase substrate. Here we describe the in vivo characterization of the novel γ-secretase inhibitor SCH 697466 in rodents. Although SCH 697466 was effective at lowering Aß, Notch-related side effects in the intestine and thymus were observed following subchronic administration at doses that provided sustained and complete lowering of Aß. However, additional studies revealed that both partial but sustained lowering of Aßand complete but less sustained lowering of Aß were successful approaches for managing Notch-related side effects. Further, changes in several Notch-related biomarkers paralleled the side effect observations. Taken together, these studies demonstrated that, by carefully varying the extent and duration of Aß lowering by γ-secretase inhibitors, it is possible to obtain robust and sustained lowering of Aß without evidence of Notch-related side effects.

17.
J Med Chem ; 55(21): 9331-45, 2012 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-22989333

RESUMEN

On the basis of our observation that the biaryl substituent of iminopyrimidinone 7 must be in a pseudoaxial conformation to occupy the contiguous S1-S3 subsites of BACE1, we have designed a novel fused bicyclic iminopyrimidinone scaffold intended to favor this bioactive conformation. Strategic incorporation of a nitrogen atom in the new constrained ring allowed us to develop SAR around the S2' binding pocket and ultimately resulted in analogues with low nanomolar potency for BACE1. In particular, optimization of the prime side substituent led to major improvements in potency by displacement of two conserved water molecules from a region near S2'. Further optimization of the pharmacokinetic properties of this fused pyrrolidine series, in conjunction with facile access to a rat pharmacodynamic model, led to identification of compound 43, which is an orally active, brain penetrant inhibitor that reduces Aß(40) in the plasma, CSF, and cortex of rats in a dose-dependent manner.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Compuestos Bicíclicos Heterocíclicos con Puentes/síntesis química , Nitrilos/síntesis química , Pirimidinas/síntesis química , Pirimidinonas/síntesis química , Tiofenos/síntesis química , Administración Oral , Secretasas de la Proteína Precursora del Amiloide/química , Péptidos beta-Amiloides/metabolismo , Animales , Ácido Aspártico Endopeptidasas/química , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacocinética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Corteza Cerebral/metabolismo , Cristalografía por Rayos X , Células HEK293 , Humanos , Macaca fascicularis , Modelos Moleculares , Conformación Molecular , Nitrilos/farmacocinética , Nitrilos/farmacología , Fragmentos de Péptidos/metabolismo , Permeabilidad , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Pirimidinonas/farmacocinética , Pirimidinonas/farmacología , Teoría Cuántica , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Termodinámica , Tiofenos/farmacocinética , Tiofenos/farmacología
18.
Neuropharmacology ; 60(4): 617-25, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21167848

RESUMEN

NMDA glutamate receptors (NMDARs) and nicotinic acetylcholine receptors (nAChRs) are both involved in learning and synaptic plasticity. Increasing evidence suggests processes mediated by these receptors may interact to modulate learning; however, little is known about the neural substrates involved in these interactive processes. The present studies investigated the effects of nicotine on MK-801 hydrogen maleate (MK-801) and DL-2-Amino-5-phosphonovaleric acid (APV)-induced disruption of contextual fear conditioning in male C57BL/6J mice, using direct drug infusion and selective nAChR antagonists to define the brain regions and the nAChR subtypes involved. Mice treated with MK-801 showed a deficit in contextual fear conditioning that was ameliorated by nicotine. Direct drug infusion demonstrated that the NMDAR antagonists disrupted hippocampal function and that nicotine acted in the dorsal hippocampus to ameliorate the deficit in learning. The high-affinity nAChR antagonist Dihydro-ß-erythroidine hydrobromide (DhßE) blocked the effects of nicotine on MK-801-induced deficits while the α7 nAChR antagonist methyllycaconitine citrate salt hydrate (MLA) did not. These results suggest that NMDARs and nAChRs may mediate similar hippocampal processes involved in contextual fear conditioning. Furthermore, these results may have implications for developing effective therapeutics for the cognitive deficits associated with schizophrenia because a large subset of patients with schizophrenia exhibit cognitive deficits that may be related to NMDAR dysfunction and smoke at much higher rates than the healthy population, which may be an attempt to ameliorate cognitive deficits.


Asunto(s)
Condicionamiento Clásico/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Miedo/efectos de los fármacos , Hipocampo/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Nicotínicos/metabolismo , Análisis de Varianza , Animales , Aprendizaje por Asociación/efectos de los fármacos , Aprendizaje por Asociación/fisiología , Condicionamiento Clásico/fisiología , Miedo/fisiología , Hipocampo/metabolismo , Masculino , Ratones , Valina/análogos & derivados , Valina/farmacología
19.
Pharmacol Biochem Behav ; 98(2): 181-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21122808

RESUMEN

Metabotropic glutamate receptor 1 (mGluR1) antagonists interfere with learning and memory; however, their role in motor function is not well elucidated despite their abundance in brain areas implicated in the control of movement. Here, the effects of mGluR1 antagonism on movement, coordination, and motor learning were investigated. JNJ16259685, a selective mGluR1 antagonist (negative allosteric modulator), was tested in assays of motor skill, and motor learning in rats and mice. JNJ16259685 produced very minimal effects on locomotor activity and posture up to a dose of 30 mg/kg. Motor skill was unaffected for well-learned tasks (up to 30 mg/kg) in rats, but impaired in mice. Both rats and mice rats were profoundly impaired (0.3 mg/kg) in the acquisition of a novel motor skill (rotarod). These results implicate the mGluR1 receptor in the acquisition of novel motor skills. JNJ16259685 dramatically reduced rearing behavior, exploration of a novel environment and lever pressing for a food reward (rat: 0.3 mg/kg; mouse: 1 mg/kg). JNJ16259685 (30 mg/kg) had no effect on reflexive startle responses to loud auditory stimuli or foot shock in mice. Previous groups have proposed that mGluR1 antagonists induce a general reduction in motivation. The effects seen here to reduce exploration and reward are consistent with that hypothesis. Pharmacological inhibition of the mGluR1 receptor has a modest effect on motor function but blocks motor learning and may reduce motivation to perform simple behaviors.


Asunto(s)
Actividad Motora/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Quinolinas/farmacología , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Animales , Antagonistas de Aminoácidos Excitadores/farmacología , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Actividad Motora/fisiología , Desempeño Psicomotor/fisiología , Ratas , Ratas Long-Evans , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA