Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Pharmacol Res ; 192: 106741, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37149116

RESUMEN

Tobacco and nicotine use are associated with disease susceptibility and progression. Health challenges associated with nicotine and smoking include developmental delays, addiction, mental health and behavioral changes, lung disease, cardiovascular disease, endocrine disorders, diabetes, immune system changes, and cancer. Increasing evidence suggests that nicotine-associated epigenetic changes may mediate or moderate the development and progression of a myriad of negative health outcomes. In addition, nicotine exposure may confer increased lifelong susceptibility to disease and mental health challenges through alteration of epigenetic signaling. This review examines the relationship between nicotine exposure (and smoking), epigenetic changes, and maladaptive outcomes that include developmental disorders, addiction, mental health challenges, pulmonary disease, cardiovascular disease, endocrine disorders, diabetes, immune system changes, and cancer. Overall, findings support the contention that nicotine (or smoking) associated altered epigenetic signaling is a contributing factor to disease and health challenges.


Asunto(s)
Nicotina , Cese del Hábito de Fumar , Nicotina/efectos adversos , Fumar/efectos adversos , Epigénesis Genética , Biología
2.
Dev Psychobiol ; 65(2): e22367, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36811365

RESUMEN

Multigenerational inheritance is a nongenomic form of heritability characterized by altered phenotypes in the first generation born from the exposed parent. Multigenerational factors may account for inconsistencies and gaps in heritable nicotine addiction vulnerability. Our lab previously found that F1 offspring of male C57BL/6J mice chronically exposed to nicotine exhibited altered hippocampus functioning and related learning, nicotine-seeking, nicotine metabolism, and basal stress hormones. In an effort to identify germline mechanisms underlying these multigenerational phenotypes, the current study sequenced small RNA extracted from sperm of males chronically administered nicotine using our previously established exposure model. We identified 16 miRNAs whose expression in sperm was dysregulated by nicotine exposure. A literature review of previous research on these transcripts suggested an enrichment for regulation of psychological stress and learning. mRNAs predicted to be regulated by differentially expressed sperm small RNAs were further analyzed using exploratory enrichment analysis, which suggested potential modulation of pathways related to learning, estrogen signaling, and hepatic disease, among other findings. Overall, our findings point to links between nicotine-exposed F0 sperm miRNA and altered F1 phenotypes in this multigenerational inheritance model, particularly F1 memory, stress, and nicotine metabolism. These findings provide a valuable foundation for future functional validation of these hypotheses and characterization of mechanisms underlying male-line multigenerational inheritance.


Asunto(s)
MicroARNs , Nicotina , Ratones , Animales , Masculino , Nicotina/metabolismo , Ratones Endogámicos C57BL , Semen , Espermatozoides/metabolismo , MicroARNs/metabolismo
3.
Eur J Neurosci ; 55(9-10): 2196-2215, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34402112

RESUMEN

Recent findings indicate that stress exposure during adolescence contributes to the development of both nicotine use and affective disorders, suggesting a potential shared biological pathway. One key system that may mediate the association between adolescent stress and nicotine or affective outcomes is the hypothalamic-pituitary-adrenal (HPA) axis. Here we reviewed evidence regarding the effects of adolescent stress on nicotine responses and affective phenotypes and the role of the HPA-axis in these relationships. Literature indicates that stress, possibly via HPA-axis dysfunction, is a risk factor for both nicotine use and affective disorders. In rodent models, adolescent stress modulates behavioural responses to nicotine and increases the likelihood of affective disorders. The exact role that the HPA-axis plays in altering nicotine sensitivity and affective disorder development after adolescent stress remains unclear. However, it appears likely that adolescent stress-induced nicotine use and affective disorders are precipitated by repetitive activation of a hyperactive HPA-axis. Together, these preclinical studies indicate that adolescent stress is a risk factor for nicotine use and anxiety/depression phenotypes. The findings summarized here suggest that the HPA-axis mediates this relationship. Future studies that pharmacologically manipulate the HPA-axis during and after adolescent stress are critical to elucidate the exact role that the HPA-axis plays in the development of nicotine use and affective disorders following adolescent stress.


Asunto(s)
Nicotina , Roedores , Animales , Sistema Hipotálamo-Hipofisario/metabolismo , Trastornos del Humor/metabolismo , Nicotina/efectos adversos , Sistema Hipófiso-Suprarrenal/metabolismo , Estrés Psicológico/metabolismo
4.
Addict Biol ; 26(1): e12859, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-31782218

RESUMEN

Nicotine use remains highly prevalent with tobacco and e-cigarette products consumed worldwide. However, increasing evidence of transgenerational epigenetic inheritance suggests that nicotine use may alter behavior and neurobiology in subsequent generations. We tested the effects of chronic paternal nicotine exposure in C57BL6/J mice on fear conditioning in F1 and F2 offspring, as well as conditioned fear extinction and spontaneous recovery, nicotine self-administration, hippocampal cholinergic functioning, RNA expression, and DNA methylation in F1 offspring. Paternal nicotine exposure was associated with enhanced contextual and cued fear conditioning and spontaneous recovery of extinguished fear memories. Further, nicotine reinforcement was reduced in nicotine-sired mice, as assessed in a self-administration paradigm. These behavioral phenotypes were coupled with altered response to nicotine, upregulated hippocampal nicotinic acetylcholine receptor binding, reduced evoked hippocampal cholinergic currents, and altered methylation and expression of hippocampal genes related to neural development and plasticity. Gene expression analysis suggests multigenerational effects on broader gene networks potentially involved in neuroplasticity and mental disorders. The changes in fear conditioning similarly suggest phenotypes analogous to anxiety disorders similar to post-traumatic stress.


Asunto(s)
Miedo/efectos de los fármacos , Hipocampo/metabolismo , Memoria/efectos de los fármacos , Nicotina/farmacología , Exposición Paterna/efectos adversos , Animales , Condicionamiento Psicológico/efectos de los fármacos , Señales (Psicología) , Extinción Psicológica , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba/efectos de los fármacos
5.
Neurobiol Learn Mem ; 172: 107234, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32428585

RESUMEN

Adolescent alcohol use is a widespread problem in the United States. In both humans and rodents, alcohol can impair learning and memory processes mediated by forebrain areas such as the prefrontal cortex (PFC) and hippocampus (HC). Adolescence is a period in which alcohol use often begins, and it is also a time that can be uniquely sensitive to the detrimental effects of alcohol. Exposure to alcohol during adolescence can cause persisting alterations in PFC and HC neurobiology that are linked to cognitive impairments, including changes in neurogenesis, inflammation, and various neurotransmitter systems in rodent models. Consistent with this, chronic adolescent alcohol exposure can cause PFC-dependent learning impairments that persist into adulthood. Deficits in adult HC-dependent learning after adolescent alcohol exposure have also been reported, but these findings are less consistent. Overall, evidence summarized in this review indicates that adolescent exposure to alcohol can produce long-term detrimental effects on forebrain-dependent cognitive processes.


Asunto(s)
Alcoholismo/fisiopatología , Alcoholismo/psicología , Hipocampo/fisiopatología , Aprendizaje , Corteza Prefrontal/fisiopatología , Consumo de Alcohol en Menores/psicología , Animales , Disfunción Cognitiva/etiología , Etanol/administración & dosificación , Hipocampo/efectos de los fármacos , Humanos , Aprendizaje/efectos de los fármacos , Ratones , Corteza Prefrontal/efectos de los fármacos , Ratas
6.
Addict Biol ; 25(3): e12769, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31099135

RESUMEN

Adolescence represents increased susceptibility to stress that increases risk for nicotine dependence. The present study examined the interactive effects of brief exposure to stress (shipping/transportation or experimentally induced) and chronic nicotine during adolescence on cognitive function and stress reactivity in adulthood. Adolescent (P31), but not young adult (P47), C57BL/6J mice had higher levels of corticosterone after shipping vs mice bred onsite. Shipped preadolescent (P23) and adolescent (P38) mice, but not those bred onsite, exposed to nicotine showed deficits in contextual fear learning when tested in adulthood. Adult learning deficits were replicated in adolescent mice bred onsite, exposed to experimentally induced stress, and administered chronic nicotine. Stress and nicotine during adolescence resulted in higher expression of hippocampal glucocorticoid receptors and corticotropin-releasing factor receptors and blunted restraint induced CORT release in adulthood. Importantly, studies examining adolescent behavior in mice should consider stress influences outcomes.


Asunto(s)
Cognición/efectos de los fármacos , Hipocampo/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Estrés Psicológico/fisiopatología , Animales , Cognición/fisiología , Corticosterona/metabolismo , Miedo , Hipocampo/fisiopatología , Aprendizaje/fisiología , Ratones , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de Glucocorticoides/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Estrés Psicológico/metabolismo
7.
Eur J Neurosci ; 50(3): 2453-2466, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-29949212

RESUMEN

Addictions are highly heritable disorders, with heritability estimates ranging from 39% to 72%. Multiple studies suggest a link between paternal drug abuse and addiction in their children. However, patterns of inheritance cannot be explained purely by Mendelian genetic mechanisms. Exposure to drugs of abuse results in epigenetic changes that may be passed on through the germline. This mechanism of epigenetic transgenerational inheritance may provide a link between paternal drug exposure and addiction susceptibility in the offspring. Recent studies have begun to investigate the effect of paternal drug exposure on behavioral and neurobiological phenotypes in offspring of drug-exposed fathers in rodent models. This review aims to discuss behavioral and neural effects of paternal exposure to alcohol, cocaine, opioids, and nicotine. Although a special focus will be on addiction-relevant behaviors, additional behavioral effects including cognition, anxiety, and depressive-like behaviors will be discussed.


Asunto(s)
Encéfalo/fisiología , Hijo de Padres Discapacitados/psicología , Drogas Ilícitas/efectos adversos , Exposición Paterna/efectos adversos , Trastornos Relacionados con Sustancias/genética , Trastornos Relacionados con Sustancias/psicología , Adulto , Encéfalo/efectos de los fármacos , Niño , Trastornos de la Conducta Infantil/diagnóstico , Trastornos de la Conducta Infantil/genética , Trastornos de la Conducta Infantil/psicología , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/fisiología , Humanos , Masculino , Trastornos Relacionados con Sustancias/diagnóstico
8.
Learn Mem ; 25(9): 481-491, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30115770

RESUMEN

During adolescence, the brain continues to undergo vital developmental processes. In turn, complex behavioral and cognitive skills emerge. Unfortunately, neurobiological development during adolescence can be influenced by environmental factors such as drug exposure. Engaging in drug use during adolescence has been a long-standing health concern, especially how it predicts or relates to drug using behavior later in life. However, recent findings suggest that other behavioral domains, such as learning and memory, are also vulnerable to adolescent drug use. Moreover, it is becoming increasingly apparent that deficits in learning and memory following adolescent drug use endure into adulthood, well after drug exposure has subsided. Although persistent effects suggest an interaction between drug exposure and ongoing development during adolescence, the exact acute and long-term consequences of adolescent drug exposure on substrates of learning and memory are not fully understood. Thus, this review will summarize human and animal findings on the enduring cognitive deficits due to adolescent drug exposure. Moreover, due to the fact that adolescents are more likely to consume drugs of abuse legally available to adults, this review will focus on alcohol, nicotine, and marijuana. Further, given the critical role of the frontal cortex and hippocampus in various learning and memory domains, the impact adolescent use of the previous listed drugs on the neurobiology within these regions will also be discussed.


Asunto(s)
Conducta del Adolescente/efectos de los fármacos , Desarrollo del Adolescente/efectos de los fármacos , Encéfalo/efectos de los fármacos , Cannabis/efectos adversos , Disfunción Cognitiva/inducido químicamente , Etanol/efectos adversos , Nicotina/efectos adversos , Trastornos Relacionados con Sustancias/complicaciones , Adolescente , Animales , Humanos
9.
Neurobiol Learn Mem ; 155: 239-248, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30099202

RESUMEN

Earlier initiation of smoking correlates with higher risk of nicotine dependence, mental health problems, and cognitive impairments. Additionally, exposure to nicotine and/or tobacco smoke during critical developmental periods is associated with lasting epigenetic modifications and altered gene expression. This study examined whether adolescent nicotine exposure alters adult hippocampus-dependent learning, involving persistent changes in hippocampal DNA methylation and if choline, a dietary methyl donor, would reverse and mitigate these alterations. Mice were chronically treated with nicotine (12.6 mg/kg/day) starting at post-natal day 23 (pre-adolescent), p38 (late adolescent), or p54 (adult) for 12 days followed by a 30-day period during which they consumed either standard chow or chow supplemented with choline (9 g/kg). Mice then were tested for fear-conditioning and dorsal hippocampi were dissected for whole genome methylation and selected gene expression analyses. Nicotine exposure starting at p21 or p38, but not p54, disrupted adult hippocampus-dependent fear conditioning. Choline supplementation ameliorated these deficits. 462 genes in adult dorsal hippocampus from mice exposed to nicotine as adolescents showed altered promoter methylation that was reversed by choline supplementation. Gene network analysis revealed that chromatin remodeling genes were the most enriched category whose methylation was altered by nicotine and reversed by choline dietary supplementation. Two key chromatin remodeling genes, Smarca2 and Bahcc1, exhibited inversely correlated changes in methylation and expression due to nicotine exposure; this was reversed by choline. Our findings support a role for epigenetic modification of hippocampal chromatin remodeling genes in long-term learning deficits induced by adolescent nicotine and their amelioration by dietary choline supplementation.


Asunto(s)
Colina/administración & dosificación , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Hipocampo/efectos de los fármacos , Nicotina/administración & dosificación , Factores de Edad , Animales , Fumar Cigarrillos/genética , Fumar Cigarrillos/psicología , Condicionamiento Clásico/fisiología , Metilación de ADN , Miedo , Hipocampo/metabolismo , Masculino , Ratones Endogámicos C57BL
10.
Nicotine Tob Res ; 20(7): 789-799, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29065200

RESUMEN

Introduction: Scientific discoveries over the past few decades have provided significant insight into the abuse liability and negative health consequences associated with tobacco and nicotine-containing products. While many of these advances have led to the development of policies and laws that regulate access to and formulations of these products, further research is critical to guide future regulatory efforts, especially as novel nicotine-containing products are introduced and selectively marketed to vulnerable populations. Discussion: In this narrative review, we provide an overview of the scientific findings that have impacted regulatory policy and discuss considerations for further translation of science into policy decisions. We propose that open, bidirectional communication between scientists and policy makers is essential to develop transformative preventive- and intervention-focused policies and programs to reduce appeal, abuse liability, and toxicity of the products. Conclusions: Through these types of interactions, collaborative efforts to inform and modify policy have the potential to significantly decrease the use of tobacco and alternative nicotine products and thus enhance health outcomes for individuals. Implications: This work addresses current topics in the nicotine and tobacco research field to emphasize the importance of basic science research and provide examples of how it can be utilized to inform public policy. In addition to relaying current thoughts on the topic from experts in the field, the article encourages continued efforts and communication between basic scientists and policy officials.


Asunto(s)
Investigación Biomédica/legislación & jurisprudencia , Nicotina , Política Pública/legislación & jurisprudencia , Productos de Tabaco/legislación & jurisprudencia , Investigación Biomédica/métodos , Humanos , Nicotina/normas , Productos de Tabaco/normas , Tabaquismo/prevención & control
11.
Neurobiol Learn Mem ; 145: 143-150, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29017931

RESUMEN

Previous research has shown that acute nicotine, an agonist of nAChRs, impaired fear extinction. However, the effects of acute nicotine on consolidation of contextual fear extinction memories and associated cell signaling cascades are unknown. Therefore, we examined the effects of acute nicotine injections before (pre-extinction) and after (post-extinction) contextual fear extinction on behavior and the phosphorylation of dorsal and ventral hippocampal ERK1/2 and JNK1 and protein levels on the 1st and 3rd day of extinction. Our results showed that acute nicotine administered prior to extinction sessions downregulated the phosphorylated forms of ERK1/2 in the ventral hippocampus, but not dorsal hippocampus, and JNK1 in both dorsal and ventral hippocampus on the 3rd extinction day. These effects were absent on the 1st day of extinction. We also showed that acute nicotine administered immediately and 30 min, but not 6 h, following extinction impaired contextual fear extinction suggesting that acute nicotine disrupts consolidation of contextual fear extinction memories. Finally, acute nicotine injections immediately after extinction sessions upregulated the phosphorylated forms of ERK1/2 in the ventral hippocampus, but did not affect JNK1. These results show that acute nicotine impairs contextual fear extinction potentially by altering molecular processes responsible for the consolidation of extinction memories.


Asunto(s)
Extinción Psicológica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Consolidación de la Memoria/efectos de los fármacos , Memoria a Largo Plazo/efectos de los fármacos , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Animales , Condicionamiento Clásico/efectos de los fármacos , Miedo , Hipocampo/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Fosforilación
13.
Nicotine Tob Res ; 19(1): 87-93, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27613891

RESUMEN

INTRODUCTION: Adolescent onset of nicotine abuse is correlated with worse chances at successful abstinence in adulthood. One reason for this may be due to enduring learning deficits resulting from nicotine use during adolescence. Previous work has indicated that chronic nicotine administration beginning in late adolescence (PND38) caused learning deficits in contextual fear when tested in adulthood. The purpose of this study was to determine if chronic nicotine treatment during adolescence would alter sensitivity to nicotine's cognitive enhancing properties in adulthood. METHODS: C57BL/6J mice received saline or chronic nicotine (12.6mg/kg/day) during adolescence (postnatal day 38) or adulthood (postnatal day 54) for a period of 12 days. Following a 30-day protracted abstinence, mice received either an acute injection of saline or nicotine (0.045, 0.18, and 0.36mg/kg) prior to training and testing a mouse model of contextual fear. RESULTS: It was found that chronic nicotine administration in adult mice did not alter sensitivity to acute nicotine following a protracted abstinence. In adolescent mice, chronic nicotine administration disrupted adult learning and decreased sensitivity to acute nicotine in adulthood as only the highest dose tested (0.36mg/kg) was able to enhance contextual fear learning. CONCLUSIONS: These results suggest that adolescent nicotine exposure impairs learning in adulthood, which could increase the risk for continued nicotine use in adulthood by requiring administration of higher doses of nicotine to reverse learning impairments caused by adolescent nicotine exposure. IMPLICATIONS: Results from this study add to the growing body of literature suggesting chronic nicotine exposure during adolescence leads to impaired learning in adulthood and demonstrates that nicotine exposure during adolescence attenuates the cognitive enhancing effects of acute nicotine in adulthood, which suggests altered cholinergic function.


Asunto(s)
Miedo/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Nicotina/administración & dosificación , Nicotina/farmacología , Tabaquismo/psicología , Factores de Edad , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Masculino , Ratones Endogámicos C57BL
14.
Learn Mem ; 23(10): 515-33, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27634143

RESUMEN

It has long been hypothesized that conditioning mechanisms play major roles in addiction. Specifically, the associations between rewarding properties of drugs of abuse and the drug context can contribute to future use and facilitate the transition from initial drug use into drug dependency. On the other hand, the self-medication hypothesis of drug abuse suggests that negative consequences of drug withdrawal result in relapse to drug use as an attempt to alleviate the negative symptoms. In this review, we explored these hypotheses and the involvement of the hippocampus in the development and maintenance of addiction to widely abused drugs such as cocaine, amphetamine, nicotine, alcohol, opiates, and cannabis. Studies suggest that initial exposure to stimulants (i.e., cocaine, nicotine, and amphetamine) and alcohol may enhance hippocampal function and, therefore, the formation of augmented drug-context associations that contribute to the development of addiction. In line with the self-medication hypothesis, withdrawal from stimulants, ethanol, and cannabis results in hippocampus-dependent learning and memory deficits, which suggest that an attempt to alleviate these deficits may contribute to relapse to drug use and maintenance of addiction. Interestingly, opiate withdrawal leads to enhancement of hippocampus-dependent learning and memory. Given that a conditioned aversion to drug context develops during opiate withdrawal, the cognitive enhancement in this case may result in the formation of an augmented association between withdrawal-induced aversion and withdrawal context. Therefore, individuals with opiate addiction may return to opiate use to avoid aversive symptoms triggered by the withdrawal context. Overall, the systematic examination of the role of the hippocampus in drug addiction may help to formulate a better understanding of addiction and underlying neural substrates.


Asunto(s)
Hipocampo/efectos de los fármacos , Drogas Ilícitas/efectos adversos , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Trastornos Relacionados con Sustancias/fisiopatología , Animales , Hipocampo/fisiopatología , Humanos , Aprendizaje/fisiología , Memoria/fisiología , Plasticidad Neuronal/fisiología
15.
Learn Mem ; 23(8): 405-14, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27421892

RESUMEN

Exposure therapy, which focuses on extinguishing fear-triggering cues and contexts, is widely used to treat post-traumatic stress disorder (PTSD). Yet, PTSD patients who received successful exposure therapy are vulnerable to relapse of fear response after a period of time, a phenomenon known as spontaneous recovery (SR). Increasing evidence suggests ventral hippocampus, basolateral amygdala, and infralimbic cortex may be involved in SR. PTSD patients also show high rates of comorbidity with nicotine dependence. While the comorbidity between smoking and PTSD might suggest nicotine may alter SR, the effects of nicotine on SR of contextual fear are unknown. In the present study, we tested the effects of acute nicotine administration on SR of extinguished contextual fear memories and c-fos immediate early gene immunohistochemistry in mice. Our results demonstrated that acute nicotine enhanced SR of extinguished fear whereas acute nicotine did not affect retrieval of unextinguished contextual memories. This suggests that the effect of acute nicotine on SR is specific for memories that have undergone extinction treatment. C-fos immunoreactive (IR) cells in the ventral hippocampus and basolateral amygdala were increased in the nicotine-treated mice following testing for SR, whereas the number of IR cells in the infralimbic cortex was decreased in the same group. Overall, this study suggests that nicotine may adversely affect context-specific relapse of fear memories and this effect is potentially mediated by the suppression of cortical regions and increased activity in the ventral hippocampus and amygdala.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Nicotina/administración & dosificación , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Condicionamiento Clásico , Señales (Psicología) , Expresión Génica , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones Endogámicos C57BL , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Proteínas Proto-Oncogénicas c-fos/biosíntesis
16.
Eur J Neurosci ; 44(10): 2818-2828, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27623427

RESUMEN

Nicotine dependence is associated with increased risk for emotional, cognitive and neurological impairments later in life. This study investigated the long-term effects of nicotine exposure during adolescence and adulthood on measures of depression, anxiety, learning and hippocampal pyramidal cell morphology. Mice (C57BL/6J) received saline or nicotine for 12 days via pumps implanted on postnatal day 32 (adolescent) or 54 (adults). Thirty days after cessation of nicotine/saline, mice were tested for learning using contextual fear conditioning, depression-like behaviors using the forced swim test or anxiety-like behaviors with the elevated plus maze. Brains from nicotine- or saline-exposed mice were processed with Golgi stain for whole neuron reconstruction in the CA1 and CA3 regions of the hippocampus. Results demonstrate higher depression-like responses in both adolescent and adult mice when tested during acute nicotine withdrawal. Heightened depression-like behaviors persisted when tested after 30 days of nicotine abstinence in mice exposed as adolescents, but not adults. Adult, but not adolescent, exposure to nicotine resulted in increased open-arm time when tested after 30 days of abstinence. Nicotine exposure during adolescence caused deficits in contextual fear learning indicated by lower levels of freezing to the context as compared with controls when tested 30 days later. In addition, reduced dendritic length and complexity in the apical CA1 branches in adult mice exposed to nicotine during adolescence were found. These results demonstrate that nicotine exposure and withdrawal can have long-term effects on emotional and cognitive functioning, particularly when nicotine exposure occurs during the critical period of adolescence.


Asunto(s)
Cognición/efectos de los fármacos , Depresión/etiología , Emociones/efectos de los fármacos , Hipocampo/efectos de los fármacos , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Animales , Condicionamiento Clásico , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Células Piramidales/citología , Células Piramidales/efectos de los fármacos
17.
Neurobiol Learn Mem ; 134 Pt B: 193-209, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27422017

RESUMEN

Translational assays of cognition that are similarly implemented in both lower and higher-order species, such as rodents and primates, provide a means to reconcile preclinical modeling of psychiatric neuropathology and clinical research. To this end, Pavlovian conditioning has provided a useful tool for investigating cognitive processes in both lab animal models and humans. This review focuses on trace conditioning, a form of Pavlovian conditioning typified by the insertion of a temporal gap (i.e., trace interval) between presentations of a conditioned stimulus (CS) and an unconditioned stimulus (US). This review aims to discuss pre-clinical and clinical work investigating the mnemonic processes recruited for trace conditioning. Much work suggests that trace conditioning involves unique neurocognitive mechanisms to facilitate formation of trace memories in contrast to standard Pavlovian conditioning. For example, the hippocampus and prefrontal cortex (PFC) appear to play critical roles in trace conditioning. Moreover, cognitive mechanistic accounts in human studies suggest that working memory and declarative memory processes are engaged to facilitate formation of trace memories. The aim of this review is to integrate cognitive and neurobiological accounts of trace conditioning from preclinical and clinical studies to examine involvement of working and declarative memory.


Asunto(s)
Condicionamiento Clásico/fisiología , Hipocampo/fisiología , Memoria a Largo Plazo/fisiología , Memoria a Corto Plazo/fisiología , Corteza Prefrontal/fisiología , Animales , Humanos
18.
Neurobiol Learn Mem ; 128: 17-22, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26688111

RESUMEN

Previously, studies from our lab have shown that while acute nicotine administered prior to training and testing enhances contextual fear conditioning, acute nicotine injections prior to extinction sessions impair extinction of contextual fear. Although there is also strong evidence showing that the acute nicotine's enhancing effects on contextual fear conditioning require high-affinity α4ß2 nicotinic acetylcholine receptors (nAChRs), it is unknown which nAChR subtypes are involved in the acute nicotine-induced impairment of contextual fear extinction. In this study, we investigated the effects of acute nicotine administration on contextual fear extinction in knock-out (KO) mice lacking α4, ß2 or α7 subtypes of nAChRs and their wild-type (WT) littermates. Both KO and WT mice were first trained and tested for contextual fear conditioning and received a daily contextual extinction session for 4 days. Subjects received intraperitoneal injections of nicotine (0.18 mg/kg) or saline 2-4 min prior to each extinction session. Our results showed that the mice that lack α4 and ß2 subtypes of nAChRs showed normal contextual fear extinction but not the acute nicotine-induced impairment while the mice that lack the α7 subtype showed both normal contextual extinction and nicotine-induced impairment of contextual extinction. In addition, control experiments showed that acute nicotine-induced impairment of contextual fear extinction persisted when nicotine administration was ceased and repeated acute nicotine administrations alone did not induce freezing behavior in the absence of context-shock learning. These results clearly demonstrate that high-affinity α4ß2 nAChRs are necessary for the effects of acute nicotine on contextual fear extinction.


Asunto(s)
Extinción Psicológica/fisiología , Miedo/fisiología , Nicotina/administración & dosificación , Receptores Nicotínicos/fisiología , Animales , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Receptores Nicotínicos/genética , Receptor Nicotínico de Acetilcolina alfa 7/genética , Receptor Nicotínico de Acetilcolina alfa 7/fisiología
19.
Neurobiol Learn Mem ; 128: 110-6, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26775017

RESUMEN

Nicotine is a major psychoactive and addictive component of tobacco. Although cessation of tobacco use produces various somatic and affective symptoms, withdrawal-related cognitive deficits are considered to be a critical symptom that predict relapse. Therefore, delineating the cognitive mechanisms of nicotine withdrawal may likely provide gainful insights into the neurobiology of nicotine addiction. The present study was designed to examine the effects of nicotine withdrawal induced by mecamylamine, a non-specific nicotinic receptor (nAChR) antagonist, on cognitive control processes in mice using an operant strategy switching task. Brain-derived neurotrophic factor (BDNF) modulates synaptic transmission in frontostriatal circuits, and these circuits are critical for executive functions. Thus, we examined the effects of mecamylamine-precipitated nicotine withdrawal on prefrontal and striatal BDNF protein expression. Mice undergoing precipitated nicotine withdrawal required more trials to attain strategy switching criterion as compared to the controls. Error analysis indicated that impaired performance in these animals was mostly related to their inability to execute the new strategy. The striatal/prefrontal BDNF ratios robustly increased following precipitated nicotine withdrawal. Moreover, higher BDNF ratios were associated with longer task acquisition. Collectively, our findings illustrate that mecamylamine-induced nicotine withdrawal disrupts cognitive control processes and that these changes are possibly linked to perturbations in frontostriatal BDNF signaling.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Función Ejecutiva/fisiología , Mecamilamina/administración & dosificación , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Antagonistas Nicotínicos/administración & dosificación , Síndrome de Abstinencia a Sustancias/metabolismo , Animales , Cognición/efectos de los fármacos , Cognición/fisiología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Función Ejecutiva/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA