Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Neurosci ; 39(10): 1842-1854, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30626700

RESUMEN

Rats trained to perform a version of the rat gambling task (rGT) in which salient audiovisual cues accompany reward delivery, similar to commercial gambling products, show greater preference for risky options. Given previous demonstrations that probabilistic reinforcement schedules can enhance psychostimulant-induced increases in accumbal DA and locomotor activity, we theorized that performing this cued task could perpetuate a proaddiction phenotype. Significantly more rats developed a preference for the risky options in the cued versus uncued rGT at baseline, and this bias was further exacerbated by cocaine self-administration, whereas the choice pattern of optimal decision-makers was unaffected. The addition of reward-paired cues therefore increased the proportion of rats exhibiting a maladaptive cognitive response to cocaine self-administration. Risky choice was not associated with responding for conditioned reinforcement or a marker of goal/sign-tracking, suggesting that reward-concurrent cues precipitate maladaptive choice via a unique mechanism unrelated to simple approach toward, or responding for, conditioned stimuli. Although "protected" from any resulting decision-making impairment, optimal decision-makers trained on the cued rGT nevertheless self-administered more cocaine than those trained on the uncued task. Collectively, these data suggest that repeated engagement with heavily cued probabilistic reward schedules can drive addiction vulnerability through multiple behavioral mechanisms. Rats trained on the cued rGT also exhibited blunted locomotor sensitization and lower basal accumbal DA levels, yet greater cocaine-induced increases in accumbal DA efflux. Gambling in the presence of salient cues may therefore result in an adaptive downregulation of the mesolimbic DA system, rendering individuals more sensitive to the deleterious effects of taking cocaine.SIGNIFICANCE STATEMENT Impaired cost/benefit decision making, exemplified by preference for the risky, disadvantageous options on the Iowa Gambling Task, is associated with greater risk of relapse and treatment failure in substance use disorder. Understanding factors that enhance preference for risk may help elucidate the neurobiological mechanisms underlying maladaptive decision making in addiction, thereby improving treatment outcomes. Problem gambling is also highly comorbid with substance use disorder, and many commercial gambling products incorporate salient win-paired cues. Here we show that adding reward-concurrent cues to a rat analog of the IGT precipitates a hypodopaminergic state, characterized by blunted accumbal DA efflux and attenuated locomotor sensitization, which may contribute to the enhanced responsivity to uncertain rewards or the reinforcing effects of cocaine we observed.


Asunto(s)
Conducta Adictiva/fisiopatología , Cocaína/administración & dosificación , Señales (Psicología) , Dopamina/metabolismo , Comportamiento de Búsqueda de Drogas/fisiología , Juego de Azar/fisiopatología , Núcleo Accumbens/fisiopatología , Recompensa , Estimulación Acústica , Animales , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Locomoción/efectos de los fármacos , Masculino , Núcleo Accumbens/efectos de los fármacos , Estimulación Luminosa , Ratas Long-Evans
2.
Brain Inj ; 34(1): 140-148, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31532706

RESUMEN

Primary Objective: Traumatic brain injury (TBI) is associated with numerous psychiatric comorbidities, and subclinical psychiatric symptoms. While many symptoms have been replicated in animal models of brain injury, a vast majority of studies utilize naïve rats as subjects, which fail to mimic the complex learning history of human patients.Methods and Procedures: In the current study, we evaluated the effects of a brain injury in animals with early exposure to uncertainty on post-injury decision-making in a probabilistic task, the rodent gambling task (RGT).Main Outcomes and Results: Exposure to uncertainty resulted in a heterogeneous sample relative to prior publications, and brain-injured rats showed no deficits in choice behavior compared to shams which contrasts with large, pervasive deficits in previously published work. However, TBI increased impulsivity and caused transient changes in behavioral variables indicative of initial motivational deficits (pellets earned, omitted responses). Notably, effects of amphetamine were similar on this heterogeneous sample of rats relative to a number of other published reports, suggesting consistent effects of gross monoaminergic manipulations on choice behavior, independent of experience.Conclusions: Going forward, translational studies need to consider the heterogeneity that exists at the clinical level and account for these problems when modeling diseases in animals.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Juego de Azar , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Toma de Decisiones , Humanos , Ratas , Ratas Long-Evans , Incertidumbre
3.
Eur J Neurosci ; 50(3): 2134-2145, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30118561

RESUMEN

Traumatic brain injury (TBI) has been linked to the development of numerous psychiatric diseases, including substance use disorder. However, it can be difficult to ascertain from clinical data whether the TBI is cause or consequence of increased addiction vulnerability. Surprisingly few studies have taken advantage of animal models to investigate the causal nature of this relationship. In terms of a plausible neurobiological mechanism through which TBI could magnify the risk of substance dependence, numerous studies indicate that TBI can cause widespread disruption to monoaminergic signaling in striatal regions, and also increases neuroinflammation. In the current study, male Long-Evans rats received either a mild or severe TBI centered over the frontal cortex via controlled cortical impact, and were subsequently trained to self-administer cocaine over 10 6-hour sessions. At the end of the study, markers of striatal dopaminergic function, and levels of inflammatory cytokine levels in the frontal lobes, were assessed via western blot and multiplex ELISA, respectively. There was significantly higher cocaine intake in a subset of animals with either mild or severe TBI. However, many animals within both TBI groups failed to acquire self-administration. Principal components analysis suggested that both dopaminergic and neuroinflammatory proteins were associated with overall cocaine intake, yet only an inflammatory component was associated with acquisition of self-administration, suggesting neuroinflammation may make a more substantial contribution to the likelihood of drug-taking. Should neuroinflammation play a causal role in mediating TBI-induced addiction risk, anti-inflammatory therapy may reduce the likelihood of substance abuse in TBI populations.


Asunto(s)
Lesiones Traumáticas del Encéfalo/fisiopatología , Cocaína/farmacología , Lóbulo Frontal/fisiopatología , Inflamación/complicaciones , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Cocaína/administración & dosificación , Modelos Animales de Enfermedad , Lóbulo Frontal/metabolismo , Masculino , Ratas Long-Evans , Ratas Sprague-Dawley , Autoadministración
4.
Behav Pharmacol ; 29(7): 638-653, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30215622

RESUMEN

Stroke is a worldwide leading cause of death and long-term disability with concurrent secondary consequences that are largely comprised of mood dysfunction, as well as sensory, motor, and cognitive deficits. This review focuses on the cognitive deficits associated with stroke specific to executive dysfunction (including decision making, working memory, and cognitive flexibility) in humans, nonhuman primates, and additional animal models. Further, we review some of the cellular and molecular underpinnings of the individual components of executive dysfunction and their neuroanatomical substrates after stroke, with an emphasis on the changes that occur during biogenic monoamine neurotransmission. We concentrate primarily on changes in the catecholaminergic (dopaminergic and noradrenergic) and serotonergic systems at the levels of neurotransmitter synthesis, distribution, reuptake, and degradation. We also discuss potential secondary stroke-related behavioral deficits (specifically, poststroke depression as well as drug-abuse potential and addiction) and their relationship with stroke-induced deficits in executive function, an especially important consideration given that the average age of the human stroke population is decreasing. In the final sections, we address pharmacological considerations for the treatment of ischemia and the subsequent functional impairment, as well as current limitations in the field of stroke and executive function research.


Asunto(s)
Monoaminas Biogénicas/uso terapéutico , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Función Ejecutiva/efectos de los fármacos , Psicofarmacología/métodos , Accidente Cerebrovascular/complicaciones , Animales , Humanos
5.
Behav Pharmacol ; 29(7): 617-637, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30215621

RESUMEN

Executive function is an umbrella term that includes cognitive processes such as decision-making, impulse control, attention, behavioral flexibility, and working memory. Each of these processes depends largely upon monoaminergic (dopaminergic, serotonergic, and noradrenergic) neurotransmission in the frontal cortex, striatum, and hippocampus, among other brain areas. Traumatic brain injury (TBI) induces disruptions in monoaminergic signaling along several steps in the neurotransmission process - synthesis, distribution, and breakdown - and in turn, produces long-lasting deficits in several executive function domains. Understanding how TBI alters monoamingeric neurotransmission and executive function will advance basic knowledge of the underlying principles that govern executive function and potentially further treatment of cognitive deficits following such injury. In this review, we examine the influence of TBI on the following measures of executive function - impulsivity, behavioral flexibility, and working memory. We also describe monoaminergic-systems changes following TBI. Given that TBI patients experience alterations in monoaminergic signaling following injury, they may represent a unique population with regard to pharmacotherapy. We conclude this review by discussing some considerations for pharmacotherapy in the field of TBI.


Asunto(s)
Monoaminas Biogénicas/uso terapéutico , Lesiones Traumáticas del Encéfalo/complicaciones , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Función Ejecutiva/efectos de los fármacos , Psicofarmacología/métodos , Humanos
6.
Brain Inj ; 28(2): 235-43, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24456061

RESUMEN

PRIMARY OBJECTIVE: To assess cognitive deficits in a rat model of brain injury. RESEARCH DESIGN: Cognitive deficits are some of the most pervasive and enduring symptoms of frontal traumatic brain injury (TBI) in human patients. In animal models, the assessment of cognitive deficits from TBI has primarily been limited to tests of spatial learning. Recently, simple discrimination performance has been shown to be sensitive to frontal brain damage. The current study provides a detailed characterization of deficits in a two-choice tone discrimination following a bilateral frontal controlled cortical impact injury. METHODS AND PROCEDURES: Rats were trained on a two-tone discrimination task in a standard operant chamber, then either a frontal brain injury was delivered or sham procedures performed. Following recovery, they were re-tested on the discrimination task and then tested on a reversal of the discrimination. MAIN OUTCOMES AND RESULTS: Frontal injury caused substantial deficits in responding and discrimination accuracy as well as an increase in side bias. CONCLUSIONS: Based on the outcomes seen in this study, discrimination and other operant tasks may provide a sensitive tool to assess the effect of therapeutic agents on cognitive deficits in animal models, which could lead to improved characterization of deficits and yield an improved assessment tool to aid in drug discovery.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Trastornos del Conocimiento/fisiopatología , Lóbulo Frontal/lesiones , Audición , Animales , Conducta Animal , Aprendizaje Discriminativo , Modelos Animales de Enfermedad , Lóbulo Frontal/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley
7.
J Neurotrauma ; 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38450560

RESUMEN

Traumatic brain injury (TBI) affects a large population, resulting in severe cognitive impairments. Although cognitive rehabilitation is an accepted treatment for some deficits, studies in patients are limited in ability to probe physiological and behavioral mechanisms. Therefore, animal models are needed to optimize strategies. Frontal TBI in a rat model results in robust and replicable cognitive deficits, making this an ideal candidate for investigating various behavioral interventions. In this study, we report three distinct frontal TBI experiments assessing behavior well into the chronic post-injury period using male Long-Evans rats. First, we evaluated the impact of frontal injury on local field potentials recorded simultaneously from 12 brain regions during a probabilistic reversal learning (PbR) task. Next, a set of rats were tested on a similar PbR task or an impulsivity task (differential reinforcement of low-rate behavior [DRL]) and half received salient cues associated with reinforcement contingencies to encourage engagement in the target behavior. After intervention on the PbR task, brains were stained for markers of activity. On the DRL task, cue relevance was decoupled from outcomes to determine if beneficial effects persisted on impulsive behavior. TBI decreased the ability to detect reinforced outcomes; this was evident in task performance and reward-feedback signals occurring at beta frequencies in lateral orbitofrontal cortex (OFC) and associated frontostriatal regions. The behavioral intervention improved flexibility and increased OFC activity. Intervention also reduced impulsivity, even after cues were decoupled, which was partially mediated by improvements in timing behavior. The current study established a platform to begin investigating cognitive rehabilitation in rats and identified a strong role for dysfunctional OFC signaling in probabilistic learning after frontal TBI.

8.
Neuropsychopharmacology ; 48(11): 1612-1622, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37142665

RESUMEN

Statistical errors in preclinical science are a barrier to reproducibility and translation. For instance, linear models (e.g., ANOVA, linear regression) may be misapplied to data that violate assumptions. In behavioral neuroscience and psychopharmacology, linear models are frequently applied to interdependent or compositional data, which includes behavioral assessments where animals concurrently choose between chambers, objects, outcomes, or types of behavior (e.g., forced swim, novel object, place/social preference). The current study simulated behavioral data for a task with four interdependent choices (i.e., increased choice of a given outcome decreases others) using Monte Carlo methods. 16,000 datasets were simulated (1000 each of 4 effect sizes by 4 sample sizes) and statistical approaches evaluated for accuracy. Linear regression and linear mixed effects regression (LMER) with a single random intercept resulted in high false positives (>60%). Elevated false positives were attenuated in an LMER with random effects for all choice-levels and a binomial logistic mixed effects regression. However, these models were underpowered to reliably detect effects at common preclinical sample sizes. A Bayesian method using prior knowledge for control subjects increased power by up to 30%. These results were confirmed in a second simulation (8000 datasets). These data suggest that statistical analyses may often be misapplied in preclinical paradigms, with common linear methods increasing false positives, but potential alternatives lacking power. Ultimately, using informed priors may balance statistical requirements with ethical imperatives to minimize the number of animals used. These findings highlight the importance of considering statistical assumptions and limitations when designing research studies.


Asunto(s)
Proyectos de Investigación , Humanos , Animales , Teorema de Bayes , Reproducibilidad de los Resultados , Simulación por Computador , Modelos Lineales
9.
Behav Neurosci ; 137(1): 15-28, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35901372

RESUMEN

The mechanisms underlying chronic psychiatric-like impairments after traumatic brain injury (TBI) are currently unknown. The goal of the present study was to assess the role of diet and the gut microbiome in psychiatric symptoms after TBI. Rats were randomly assigned to receive a high-fat diet (HFD) or calorie-matched low-fat diet (LFD). After 2 weeks of free access, rats began training on the rodent gambling task (RGT), a measure of risky decision-making and motor impulsivity. After training, rats received a bilateral frontal TBI or a sham procedure and continued postinjury testing for 10 weeks. Fecal samples were collected before injury and 3-, 30-, and 60 days postinjury to evaluate the gut microbiome. HFD altered the microbiome, but ultimately had low-magnitude effects on behavior and did not modify functional outcomes after TBI. Injury-induced functional deficits were far more robust; TBI substantially decreased optimal choice and increased suboptimal choice and motor impulsivity on the RGT. TBI also affected the microbiome, and a model comparison approach revealed that bacterial diversity measured 3 days postinjury was predictive of chronic psychiatric-like deficits on the RGT. A functional metagenomic analysis identified changes to dopamine and serotonin synthesis pathways as a potential candidate mechanism. Thus, the gut may be a potential acute treatment target for psychiatric symptoms after TBI, as well as a biomarker for injury and deficit severity. However, further research will be needed to confirm and extend these findings. (PsycInfo Database Record (c) 2023 APA, all rights reserved).


Asunto(s)
Lesiones Traumáticas del Encéfalo , Juego de Azar , Microbioma Gastrointestinal , Ratas , Masculino , Animales , Ratas Long-Evans , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/microbiología , Conducta Impulsiva
10.
Physiol Behav ; 257: 113998, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36257462

RESUMEN

Increased neuroinflammation relative to controls is observed in major depression. Moreover, depressive disorders are significantly elevated in conditions which increase neuroinflammation (e.g., brain injury, Parkinson's disease, Alzheimer's disease). To better understand the relationship between neuroinflammation and depression, additional research is needed. The current set of studies made use of the progressive ratio (PR) task in male rats, a stable measure of motivation which can be evaluated daily and thus is ideally suited for examining a potential role for chronic neuroinflammation in depressive-like behavior. Lipopolysaccharide (LPS) was used to induce an inflammatory response. Experiment 1 confirmed prior acute LPS administration experiments for sensitivity of the PR task, with a large effect at 2 mg/kg, a partial effect at 1 mg/kg, and no effect at 0.5 mg/kg. Experiment 2 evaluated a dose-response of continuous s.c. LPS infusion but found no significant elevation in brain cytokines after 14 days at any doses of 0.1, 0.5, 1, or 2 mg/kg/week. Experiment 3 assessed motivation during continuous s.c. infusion of a large 5 mg/kg/week LPS dose and found no significant impairments in motivation, but transient decreases in rates of lever pressing (i.e., only motoric deficits). Experiment 4 measured motivation during continuous ICV infusion of 10.5 µg/kg/week LPS and found significantly decreased motivation without changes to rates of lever pressing (i.e., only motivational deficits). Together these results suggest that the PR task is efficient for evaluating models of chronic inflammation, and that the adaptive response to chronic LPS exposure, even when delivered centrally, may necessitate alternative strategies for generating long-term neuroinflammation.


Asunto(s)
Lipopolisacáridos , Motivación , Animales , Ratas , Masculino , Lipopolisacáridos/toxicidad , Inflamación/inducido químicamente , Citocinas/metabolismo , Encéfalo/metabolismo
11.
Biomedicines ; 10(10)2022 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-36289877

RESUMEN

Pigs are becoming more common research models due to their utility in studying neurological conditions such as traumatic brain injury, Alzheimer's disease, and Huntington's Disease. However, behavioral tasks often require a large apparatus and are not automated, which may disinterest researchers in using important functional measures. To address this, we developed a touchscreen that pigs could be trained on for behavioral testing. A rack-mounted touchscreen monitor was placed in an enclosed, weighted audio rack. A pellet dispenser was operated by a radio frequency transceiver to deliver fruit-flavored sugar pellets from across the testing room. Programs were custom written in Python and executed on a microcomputer. A behavioral shaping program was designed to train pigs to interact with the screen and setup responses for future tasks. Pigs rapidly learned to interact with the screen. To demonstrate efficacy in more complex behavior, two pigs were trained on a delay discounting tasks and two pigs on a color discrimination task. The device held up to repeated testing of large pigs and could be adjusted to the height of minipigs. The device can be easily recreated and constructed at a relatively low cost. Research topics ranging from brain injury to pharmacology to vision could benefit from behavioral tasks designed to specifically interrogate relevant function. More work will be needed to develop tests which are of specific relevance to these disciplines.

12.
Front Behav Neurosci ; 16: 837654, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35548692

RESUMEN

Decision-making is substantially altered after brain injuries. Patients and rats with brain injury are more likely to make suboptimal, and sometimes risky choices. Such changes in decision-making may arise from alterations in how sensitive individuals are to outcomes. To assess this, we compiled and harmonized a large dataset from four studies of TBI, each of which evaluated behavior on the Rodent Gambling Task (RGT). We then determined whether the following were altered: (1) sensitivity to overall contingencies, (2) sensitivity to immediate outcomes, or (3) general choice phenotypes. Overall sensitivity was evaluated using the matching law, immediate sensitivity by looking at the probability of switching choices given a win or loss, and choice phenotypes by k-means clustering. We found significant reductions in sensitivity to the overall outcomes and a bias toward riskier alternatives in TBI rats. However, the substantial individual variability led to poor overall fits in matching analyses. We also found that TBI caused a significant reduction in the tendency to repeatedly choose a given option, but no difference in win- or loss-specific sensitivity. Finally, clustering revealed 5 distinct decision-making phenotypes and TBI reduced membership in the "optimal" type. The current findings support a hypothesis that TBI reduces sensitivity to contingencies. However, in the case of tasks such as the RGT, this is not a simple shift to indiscriminate or less discriminate responding. Rather, TBI rats are more likely to develop suboptimal preferences and frequently switch choices. Treatments will have to consider how this behavior might be corrected.

13.
Front Behav Neurosci ; 16: 806598, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35185489

RESUMEN

Cognitive impairment is a common symptom after traumatic brain injury (TBI). Memory, in particular, is often disrupted during chronic post-injury recovery. To understand the sex-specific effects of brain injury on retrograde and anterograde memory, we examined paired associate learning (PAL), spatial learning and memory, and fear memory after lateral fluid percussion TBI. We hypothesized that male and female mice would display unique memory deficits after TBI. PAL task acquisition was initiated via touchscreen operant conditioning 22 weeks before sham injury or TBI. Post-injury PAL testing occurred 7 weeks post-injury. Barnes maze and fear conditioning were completed at 14- and 15-weeks post-injury, respectively. Contrary to our expectations, behavioral outcomes were not primarily influenced by TBI. Instead, sex-specific differences were observed in all tasks which exposed task-specific trends in male TBI mice. Male mice took longer to complete the PAL task, but this was not affected by TBI and did not compromise the ability to make a correct choice. Latency to reach the goal box decreased across testing days in Barnes maze, but male TBI mice lagged in improvement compared to all other groups. Use of two learning indices revealed that male TBI mice were deficient in transferring information from 1 day to the next. Finally, acquisition and contextual retention of fear memory were similar between all groups. Cued retention of the tone-shock pairing was influenced by both injury and sex. Male sham mice displayed the strongest cued retention of fear memory, evidenced by increased freezing behavior across the test trial. In contrast, male TBI mice displayed reduced freezing behavior with repetitive tone exposure. An inverse relationship in freezing behavior to tone exposure was detected between female sham and TBI mice, although the difference was not as striking. Together, these studies show that retrograde memory is intact after lateral TBI. However, male mice are more vulnerable to post-injury anterograde memory deficits. These behaviors were not associated with gross pathological change near the site injury or in subcortical brain regions associated with memory formation. Future studies that incorporate pre- and post-injury behavioral analysis will be integral in defining sex-specific memory impairment after TBI.

14.
Front Behav Neurosci ; 16: 809249, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35359588

RESUMEN

Repetitive mild traumatic brain injury, or concussion, can lead to the development of long-term psychiatric impairments. However, modeling these deficits is challenging in animal models and necessitates sophisticated behavioral approaches. The current set of studies were designed to evaluate whether a rubberized versus metal impact tip would cause functional deficits, the number of injuries required to generate such deficits, and whether different psychiatric domains would be affected. Across two studies, male rats were trained in either the 5-choice serial reaction time task (5CSRT; Experiment 1) to assess attention and motor impulsivity or concurrently on the 5CSRT and the delay discounting task (Experiment 2) to also assess choice impulsivity. After behavior was stable, brain injuries were delivered with the Closed-head Injury Model of Engineered Rotational Acceleration (CHIMERA) either once per week or twice per week (Experiment 1) or just once per week (Experiment 2). Astrocyte and microglia pathology was also assayed in relevant regions of interest. CHIMERA injury caused attentional deficits across both experiments, but only increased motor impulsivity in Experiment 1. Surprisingly, choice impulsivity was actually reduced on the Delay Discounting Task after repeat injuries. However, subsequent analyses suggested potential visual issues which could alter interpretation of these and attentional data. Subtle changes in glial pathology immediately after the injury (Experiment 1) were attenuated after 4 weeks recovery (Experiment 2). Given the heterogenous findings between experiments, additional research is needed to determine the root causes of psychiatric disturbances which may arise as a results of repeated brain injuries.

15.
Curr Opin Behav Sci ; 31: 42-47, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32864398

RESUMEN

In the past several years, there has been an explosion of interest in animal models of risk-based decision-making, a fundamental process associated with gambling disorder. While early work focused on establishing various tasks for assaying decision-making, current studies are determining the (subtle and not-so-subtle) influence of cues in driving risky decisions to better understand problem gambling. In addition, these decision-making paradigms are now being used to investigate comorbid conditions such as substance dependence or brain injury and replicating observations from human patients. These animal models have now developed to a point where therapeutic interventions may be assessed for not just gambling disorder, but also a number of other conditions which engender risky decision-making.

16.
Behav Brain Res ; 395: 112879, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32841610

RESUMEN

Depression is the most common psychiatric comorbidity to be diagnosed following traumatic brain injury (TBI). In clinical populations, TBI-induced depression may be particularly difficult to treat due to both unique underlying causes and the propensity for treatment resistance. Preclinical assays are needed to characterize depressive-like behavior in models of TBI and evaluate treatments. In the current study, two traditionally-acute assays of depressive-like behaviors, the Forced Swim Task and Saccharin Preference, were extended longitudinally to evaluate chronic TBI-induced depressive-like behaviors in male rats. Two chronic measures of motivation, the Progressive Ratio (PR) task and Effort Discounting Task (EDT), were also tested. The PR measures motivation to exert effort, while the EDT parametrically evaluates choice between low- and high-effort requirements. The EDT was the only assay which captured chronic depressive-like behavior after TBI, albeit with a degree of recovery over time. We found that traditionally-acute measures (Forced Swim Task, Saccharin Preference), and even our other chronic measure (PR), failed to capture long-term deficits. We also challenged serotonin and dopamine systems (via fluoxetine and bupropion) to evaluate how TBI-induced changes to these systems might drive depressive-like behaviors. Although we found no effect of fluoxetine, high-dose bupropion differentially impaired TBI rats. These findings suggest that (1) TBI-induced depressive symptoms remain difficult to measure at the preclinical level, (2) treatment for TBI-induced depression requires further exploration, and (3) obstacles at the preclinical level may translate to treatment failure at the clinical level.


Asunto(s)
Lesiones Traumáticas del Encéfalo/psicología , Depresión/diagnóstico , Depresión/etiología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Masculino , Motivación/fisiología , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Ratas , Ratas Long-Evans
17.
Brain Res ; 1748: 147084, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32871136

RESUMEN

Traumatic brain injury affects millions of people each year and is an established risk factor for addiction. Recent animal studies have causally demonstrated that injuries can increase drug self-administration across a variety of substances. One potential behavioral mediator for this finding is an increased responsivity to drug-associated cues. This endophenotype can be identified by profiling non-drug-related behaviors. The current study evaluated several paradigms (conditioned approach, conditioned reinforcement, extinction from variable interval responding, conditioned facilitation) to determine how rats with a frontal TBI differed in their response to Pavlovian conditioning in response to food-paired cues. Surprisingly, rats with a TBI demonstrated increased goal-tracking in a conditioned approach paradigm and exerted less effort for a conditioned reinforcer. Moreover, they had slightly facilitated extinction (as demonstrated by significantly larger interresponse times) in the face of reinforcer-associated cues. Despite these effects, TBI rats still demonstrated conditioned facilitation to an auditory stimulus. Together, these effects suggest a phenotype in the opposite direction of what might be anticipated. Cues still served a strong discriminative function and altered behavior; however, they did not function as strong conditioned reinforcers for TBI animals. One potential reason for this is that substantial changes to the dopamine system after TBI may reduce the conditioned reinforcing effects of cues, but sensitize the brain to potent drugs of abuse. More research will be needed to determine whether this is the case.


Asunto(s)
Lesiones Traumáticas del Encéfalo/fisiopatología , Condicionamiento Clásico/fisiología , Refuerzo en Psicología , Animales , Señales (Psicología) , Ambiente , Masculino , Motivación/fisiología , Ratas , Ratas Long-Evans
18.
Exp Neurol ; 327: 113217, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32014440

RESUMEN

Traumatic brain injury (TBI) affects millions of individuals every year. Many of these injuries lead to lasting effects, particularly impairments in domains broadly classified as executive functions, such as impulse control and decision-making. While these impairments have been historically associated with frontal brain damage, other injuries such as concussion or parietal injury also contribute to similar dysfunction. However, it is unknown whether animal models of TBI would replicate these broad effects that are observed in human patients. In the current study, we delivered a unilateral parietal controlled cortical impact injury and assessed the performance of rats on a motoric task (rotarod) and a test of decision-making and impulsivity (rodent gambling task). TBI rats demonstrated significant motor impairments on the rotarod task; however, this did not extend to difficulties inhibiting motor actions (impulsivity). In addition, TBI caused chronic alterations to risk-based decision-making, extending out to 12 weeks post-injury. Specifically, rats with TBI preferred the riskiest, and most suboptimal option over all others. The current data suggest that models of unilateral TBI are sufficient for replicating some aspects of executive dysfunction (risky decision-making), while others are limited to frontal damage (impulsivity). These models may be used to develop therapeutics targeted at the chronic post-injury period when these symptoms often manifest in patients, a critically understudied area in preclinical TBI research.


Asunto(s)
Conducta Animal/fisiología , Lesiones Encefálicas/fisiopatología , Conducta Impulsiva/fisiología , Lóbulo Parietal/lesiones , Animales , Toma de Decisiones , Función Ejecutiva/fisiología , Masculino , Ratas , Ratas Long-Evans , Asunción de Riesgos , Prueba de Desempeño de Rotación con Aceleración Constante
19.
Behav Brain Res ; 356: 408-414, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30213663

RESUMEN

Traumatic brain injury (TBI) affects over 2.8 million people annually, and has been shown to increase motor impulsivity in both humans and animals. However, the root cause of this behavioral disinhibition is not fully understood. The goal of the current study was to evaluate whether timing behavior is disrupted after TBI, which could potentially explain increases in impulsive responding. Twenty-one male three-month old Long-Evans rats were trained on a fixed interval-18 s schedule. Following training, rats were placed on the Peak Interval Procedure, with intermittent peak trials. On peak trials, no behaviors were reinforced and response rates were recorded to determine timing ability. After reaching a stable baseline, rats received bilateral frontal TBI (n = 12) using controlled cortical impact or sham procedures (n = 9). After one week recovery, rats were re-assessed on the Peak Procedure for six weeks. An amphetamine challenge was carried out after behavior reached stable post-injury performance. TBI caused a chronic decrease/acceleration in peak time, increase in response variability, and reduction in response rate. The shifted peak time suggests that altered perception of time may contribute to impairments in response inhibition after TBI. Amphetamine significantly increased response variability, with TBI animals demonstrating greater sensitivity, but did not affect peak time in either group. These data suggest that timing may not be the sole factor explaining impulsive action after TBI given that amphetamine reduced motor impulsivity in prior studies. Further investigations will be needed to dissociate the effects of amphetamine on TBI with regard to timing behavior.


Asunto(s)
Conducta Animal/fisiología , Lesiones Encefálicas/fisiopatología , Conducta Impulsiva/fisiología , Motivación/fisiología , Animales , Encéfalo/fisiopatología , Masculino , Ratas Long-Evans , Refuerzo en Psicología , Factores de Tiempo
20.
J Neurotrauma ; 36(19): 2827-2830, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31072218

RESUMEN

Traumatic brain injury (TBI) often results in chronic psychiatric-like symptoms. In a condition with few therapeutic options, neuromodulation has emerged as a promising potential treatment avenue for these individuals. The goal of the current study was to determine if transcranial direct-current stimulation (tDCS) could treat deficits of impulsivity and attention in rats. This could then be used as a model to investigate treatment parameters and the mechanism of action underlying therapeutic effects. Rats were trained on a task to measure attention and motor impulsivity (five-choice serial reaction time task), then given a frontal, controlled cortical impact injury. After rats recovered to a new baseline, tDCS (cathodal, 10 min, 800 µA) was delivered daily prior to testing in a counterbalanced, cross-over design. Treatment with tDCS selectively reduced impulsivity in the TBI group, and the greatest recovery occurred in the rats with the largest deficits. With these data, we have established a rat model for studying the effects of tDCS on psychiatric-like dysfunction. More research is needed to determine the mechanism of action by which tDCS-related gains occur.


Asunto(s)
Conducta Animal/fisiología , Lesiones Traumáticas del Encéfalo/fisiopatología , Conducta Impulsiva/fisiología , Estimulación Transcraneal de Corriente Directa , Animales , Atención/fisiología , Ratas , Tiempo de Reacción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA