Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Neurobiol Dis ; 177: 105967, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36535550

RESUMEN

Epidemiological studies identified alcohol use disorder (AUD) as a risk factor for Alzheimer's disease (AD), yet there is conflicting evidence on how alcohol use promotes AD pathology. In this study, a 10-week moderate two-bottle choice drinking paradigm was used to identify how chronic ethanol exposure alters amyloid-ß (Aß)-related pathology, metabolism, and behavior. Ethanol-exposed APPswe/PSEN1dE9 (APP/PS1) mice showed increased brain atrophy and an increased number of amyloid plaques. Further analysis revealed that ethanol exposure led to a shift in the distribution of plaque size in the cortex and hippocampus. Ethanol-exposed mice developed a greater number of smaller plaques, potentially setting the stage for increased plaque proliferation in later life. Ethanol drinking APP/PS1 mice also exhibited deficits in nest building, a metric of self-care, as well as increased locomotor activity and central zone exploration in an open field test. Ethanol exposure also led to a diurnal shift in feeding behavior which was associated with changes in glucose homeostasis and glucose intolerance. Complementary in vivo microdialysis experiments were used to measure how acute ethanol directly modulates Aß in the hippocampal interstitial fluid (ISF). Acute ethanol transiently increased hippocampal ISF glucose levels, suggesting that ethanol directly affects cerebral metabolism. Acute ethanol also selectively increased ISF Aß40, but not ISF Aß42, levels during withdrawal. Lastly, chronic ethanol drinking increased N-methyl-d-aspartate receptor (NMDAR) and decreased γ-aminobutyric acid type-A receptor (GABAAR) mRNA levels, indicating a potential hyperexcitable shift in the brain's excitatory/inhibitory (E/I) balance. Collectively, these experiments suggest that ethanol may increase Aß deposition by disrupting metabolism and the brain's E/I balance. Furthermore, this study provides evidence that a moderate drinking paradigm culminates in an interaction between alcohol use and AD-related phenotypes with a potentiation of AD-related pathology, behavioral dysfunction, and metabolic impairment.


Asunto(s)
Enfermedad de Alzheimer , Animales , Ratones , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Modelos Animales de Enfermedad , Etanol/toxicidad , Glucosa/metabolismo , Hipocampo/metabolismo , Ratones Transgénicos , Placa Amiloide/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo
2.
Eur J Neurosci ; 57(8): 1241-1259, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36840503

RESUMEN

Alcohol use disorder (AUD) and anxiety/stressor disorders frequently co-occur and this dual diagnosis represents a major health and economic problem worldwide. The basolateral amygdala (BLA) is a key brain region that is known to contribute to the aetiology of both disorders. Although many studies have implicated BLA hyperexcitability in the pathogenesis of AUD and comorbid conditions, relatively little is known about the specific efferent projections from this brain region that contribute to these disorders. Recent optogenetic studies have shown that the BLA sends a strong monosynaptic excitatory projection to the ventral hippocampus (vHC) and that this circuit modulates anxiety- and fear-related behaviours. However, it is not known if this pathway influences alcohol drinking-related behaviours. Here, we employed a rodent operant self-administration regimen that procedurally separates appetitive (e.g. seeking) and consummatory (e.g., drinking) behaviours, chemogenetics and brain region-specific microinjections, to determine if BLA-vHC circuitry influences alcohol and sucrose drinking-related measures. We first confirmed prior optogenetic findings that silencing this circuit reduced anxiety-like behaviours on the elevated plus maze. We then demonstrated that inhibiting the BLA-vHC pathway significantly reduced appetitive drinking-related behaviours for both alcohol and sucrose while having no effect on consummatory measures. Taken together, these findings provide the first indication that the BLA-vHC circuit may regulate appetitive reward seeking directed at alcohol and natural rewards and add to a growing body of evidence suggesting that dysregulation of this pathway may contribute to the pathophysiology of AUD and anxiety/stressor-related disorders.


Asunto(s)
Alcoholismo , Complejo Nuclear Basolateral , Humanos , Hipocampo , Etanol/farmacología , Consumo de Bebidas Alcohólicas , Sacarosa/farmacología
3.
Mol Psychiatry ; 26(6): 2350-2362, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33432187

RESUMEN

Rapid antidepressants are novel treatments for major depressive disorder (MDD) and work by blocking N-methyl-D-aspartate receptors (NMDARs), which, in turn, activate the protein synthesis pathway regulated by mechanistic/mammalian target of rapamycin complex 1 (mTORC1). Our recent work demonstrates that the RNA-binding protein Fragile X Mental Retardation Protein (FMRP) is downregulated in dendrites upon treatment with a rapid antidepressant. Here, we show that the behavioral effects of the rapid antidepressant Ro-25-6981 require FMRP expression, and treatment promotes differential mRNA binding to FMRP in an mTORC1-dependent manner. Further, these mRNAs are identified to regulate transsynaptic signaling. Using a novel technique, we show that synapse formation underlying the behavioral effects of Ro-25-6981 requires GABABR-mediated mTORC1 activity in WT animals. Finally, we demonstrate that in an animal model that lacks FMRP expression and has clinical relevance for Fragile X Syndrome (FXS), GABABR activity is detrimental to the effects of Ro-25-6981. These effects are rescued with the combined therapy of blocking GABABRs and NMDARs, indicating that rapid antidepressants alone may not be an effective treatment for people with comorbid FXS and MDD.


Asunto(s)
Trastorno Depresivo Mayor , Síndrome del Cromosoma X Frágil , Animales , Antidepresivos/farmacología , Trastorno Depresivo Mayor/tratamiento farmacológico , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Síndrome del Cromosoma X Frágil/genética , Humanos , Sinapsis
4.
Mol Biol Rep ; 48(4): 3871-3876, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33880672

RESUMEN

Microtubules (MTs) are structural units in the cytoskeleton. In brain cells they are responsible for axonal transport, information processing, and signaling mechanisms. Proper function of these processes is critical for healthy brain functions. Alcohol and substance use disorders (AUD/SUDs) affects the function and organization of MTs in the brain, making them a potential neuroimaging marker to study the resulting impairment of overall neurobehavioral and cognitive processes. Our lab reported the first brain-penetrant MT-tracking Positron Emission Tomography (PET) ligand [11C]MPC-6827 and demonstrated its in vivo utility in rodents and non-human primates. To further explore the in vivo imaging potential of [11C]MPC-6827, we need to investigate its mechanism of action. Here, we report preliminary in vitro binding results in SH-SY5Y neuroblastoma cells exposed to ethanol (EtOH) or cocaine in combination with multiple agents that alter MT stability. EtOH and cocaine treatments increased MT stability and decreased free tubulin monomers. Our initial cell-binding assay demonstrated that [11C]MPC-6827 may have high affinity to free/unbound tubulin units. Consistent with this mechanism of action, we observed lower [11C]MPC-6827 uptake in SH-SY5Y cells after EtOH and cocaine treatments (e.g., fewer free tubulin units). We are currently performing in vivo PET imaging and ex vivo biodistribution studies in rodent and nonhuman primate models of AUD and SUDs and Alzheimer's disease.


Asunto(s)
Cocaína/farmacología , Etanol/farmacología , Quinazolinas/farmacología , Radiofármacos/farmacología , Radioisótopos de Carbono , Línea Celular Tumoral , Fármacos del Sistema Nervioso Central/farmacología , Humanos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Unión Proteica , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/farmacología
5.
Alcohol Clin Exp Res ; 40(6): 1202-14, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27154240

RESUMEN

Individuals diagnosed with anxiety-related illnesses are at increased risk of developing alcoholism, exhibit a telescoped progression of this disease and fare worse in recovery, relative to alcoholics that do not suffer from a comorbid anxiety disorder. Similarly, preclinical evidence supports the notion that stress and anxiety represent major risk factors for the development of alcohol use disorder (AUD). Despite the importance of understanding the link between anxiety and alcoholism, much remains unknown about the neurobiological substrates underlying this relationship. One stumbling block has been the lack of animal models that reliably reproduce the spectrum of behaviors associated with increased vulnerability to these diseases. Here, we review the literature that has examined the behavioral and neurobiological outcomes of a simple rodent adolescent social isolation procedure and discuss its validity as a model of vulnerability to comorbid anxiety disorders and alcoholism. Recent studies have provided strong evidence that adolescent social isolation of male rats leads to the expression of a variety of behaviors linked with increased vulnerability to anxiety and/or AUD, including deficits in sensory gating and fear extinction, and increases in anxiety measures and ethanol drinking. Neurobiological studies are beginning to identify mesolimbic adaptations that may contribute to the behavioral phenotype engendered by this model. Some of these changes include increased excitability of ventral tegmental area dopamine neurons and pyramidal cells in the basolateral amygdala and significant alterations in baseline and stimulated catecholamine signaling. A growing body of evidence suggests that adolescent social isolation may represent a reliable rodent model of heightened vulnerability to anxiety disorders and alcoholism in male rats. These studies provide initial support for the face, construct, and predictive validity of this model and highlight its utility in identifying neurobiological adaptations associated with increased risk of developing these disorders.


Asunto(s)
Alcoholismo/epidemiología , Alcoholismo/fisiopatología , Trastornos de Ansiedad/epidemiología , Encéfalo/fisiología , Aislamiento Social/psicología , Alcoholismo/psicología , Animales , Trastornos de Ansiedad/psicología , Comorbilidad , Modelos Animales de Enfermedad , Humanos
6.
Synapse ; 69(8): 385-95, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25963724

RESUMEN

Adolescent social isolation (SI) results in numerous behavioral alterations associated with increased risk of alcoholism. Notably, many of these changes involve the basolateral amygdala (BLA), including increased alcohol seeking. The BLA sends a strong glutamatergic projection to the nucleus accumbens and activation of this pathway potentiates reward-seeking behavior. Dopamine (DA) and norepinephrine (NE) exert powerful excitatory and inhibitory effects on BLA activity and chronic stress can disrupt the excitation-inhibition balance maintained by these catecholamines. Notably, the impact of SI on BLA DA and NE neurotransmission is unknown. Thus the aim of this study was to characterize SI-mediated catecholamine alterations in the BLA. Male Long Evans rats were housed in groups of four (GH) or in SI for 6 weeks during adolescence. DA and NE transporter levels were then measured using Western blot hybridization and baseline and ethanol-stimulated DA and NE levels were quantified using microdialysis. DA transporter levels were increased and baseline DA levels were decreased in SI compared to GH rats. SI also increased DA responses to an acute ethanol (2 g kg(-1)) challenge. While no group differences were noted in NE transporter or baseline NE levels, acute ethanol (2 g kg(-1)) only significantly increased NE levels in SI animals. Collectively, these SI-dependent changes in BLA catecholamine signaling may lead to an increase in BLA excitability and a strengthening of the glutamatergic projection between the BLA and NAc. Such changes may promote the elevated ethanol drinking behavior observed in rats subjected to chronic adolescent stress.


Asunto(s)
Complejo Nuclear Basolateral/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Dopamina/metabolismo , Etanol/farmacología , Norepinefrina/metabolismo , Aislamiento Social , Animales , Complejo Nuclear Basolateral/crecimiento & desarrollo , Complejo Nuclear Basolateral/metabolismo , Western Blotting , Cromatografía Líquida de Alta Presión , Enfermedad Crónica , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Masculino , Microdiálisis , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Distribución Aleatoria , Ratas Long-Evans
7.
J Neurosci ; 33(11): 4834-42, 2013 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-23486954

RESUMEN

Although alcoholism is a worldwide problem resulting in millions of deaths, only a small percentage of alcohol users become addicted. The specific neural substrates responsible for individual differences in vulnerability to alcohol addiction are not known. In this study, we used rodent models to study behavioral and synaptic correlates related to individual differences in the development of ethanol locomotor sensitization, a form of drug-dependent behavioral plasticity associated with addiction vulnerability. Male Swiss Webster mice were treated daily with saline or 1.8 g/kg ethanol for 21 d. Locomotor activity tests were performed once a week for 15 min immediately after saline or ethanol injections. After at least 11 d of withdrawal, cohorts of saline- or ethanol-treated mice were used to characterize the relationships between locomotor sensitization, ethanol drinking, and glutamatergic synaptic transmission in the nucleus accumbens. Ethanol-treated mice that expressed locomotor sensitization to ethanol drank significantly more ethanol than saline-treated subjects and ethanol-treated animals resilient to this form of behavioral plasticity. Moreover, ethanol-sensitized mice also had reduced accumbal NMDA receptor function and expression, as well as deficits in NMDA receptor-dependent long-term depression in the nucleus accumbens core after a protracted withdrawal. These findings suggest that disruption of accumbal core NMDA receptor-dependent plasticity may represent a synaptic correlate associated with ethanol-induced locomotor sensitization and increased propensity to consume ethanol.


Asunto(s)
Intoxicación Alcohólica/patología , Depresores del Sistema Nervioso Central/administración & dosificación , Etanol/administración & dosificación , Locomoción/fisiología , Núcleo Accumbens/patología , Receptores de N-Metil-D-Aspartato/metabolismo , Intoxicación Alcohólica/etiología , Análisis de Varianza , Animales , Bicuculina/farmacología , Biofisica , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Antagonistas de Receptores de GABA-A/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Locomoción/efectos de los fármacos , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/fisiología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Técnicas de Placa-Clamp , Autoadministración , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
8.
Alcohol Clin Exp Res ; 38(8): 2199-207, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25092210

RESUMEN

BACKGROUND: Clinically, early life stress and anxiety disorders are associated with increased vulnerability for alcohol use disorders. In male rats, early life stress, imparted by adolescent social isolation, results in long-lasting increases in a number of behavioral risk factors for alcoholism, including greater anxiety-like behaviors and ethanol (EtOH) intake. Several recent studies have begun to use this model to gain insight into the relationships among anxiety measures, stress, EtOH intake, and neurobiological correlates driving these behaviors. As prior research has noted significant sex differences in the impact of adolescent stress on anxiety measures and EtOH drinking, the current study was conducted to determine if this same model produces an "addiction vulnerable" phenotype in female rodents. METHODS: Female Long Evans rats were socially isolated (SI; 1/cage) or group housed (GH; 4/cage) for 6 weeks during adolescence. After this housing manipulation, behavioral assessment was conducted using the elevated plus maze, response to novelty in an open field environment, and the light/dark box. After behavioral testing, home cage EtOH drinking was assessed across an 8-week period. RESULTS: No group differences were detected in any of the behavioral measures of unconditioned anxiety-like behavior. Greater EtOH intake and preference were observed in SI females but these differences did not persist. CONCLUSIONS: The SI/GH model, which results in robust and enduring increases in anxiety measures and EtOH self-administration in male Long Evans rats, did not result in similar behavioral changes in female rats. These data, and that of others, suggest that adolescent social isolation is not a useful model with which to study neurobiological substrates linking antecedent anxiety and addiction vulnerability in female rats. Given the compelling epidemiological evidence that the relationship between chronic adolescent stress and alcohol addiction is particularly strong in women, there is clearly an urgent need to identify a more effective model with which to study these clinically important relationships in female rodents.


Asunto(s)
Consumo de Bebidas Alcohólicas/psicología , Ansiedad/psicología , Aislamiento Social/psicología , Animales , Modelos Animales de Enfermedad , Femenino , Vivienda para Animales , Ratas , Ratas Long-Evans , Caracteres Sexuales
9.
Alcohol Clin Exp Res ; 38(11): 2770-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25421514

RESUMEN

BACKGROUND: Early-life stress is associated with increased vulnerability to alcohol addiction. However, the neural substrates linking chronic childhood/adolescent stress and increased risk of alcohol addiction are not well understood. In the nucleus accumbens (NAc), dopamine (DA) and norepinephrine (NE) signaling can be profoundly influenced by stress, anxiety, and drugs of abuse, including ethanol (EtOH). Here, we employed a rodent model of early-life stress that results in enduring increases in behavioral risk factors of alcoholism to gain a better understanding of how chronic adolescent stress may impact the EtOH sensitivity of DA and NE release in the NAc. METHODS: Male Long-Evans rats were either group housed (GH; 4 rats/cage) or socially isolated (SI; 1 rat/cage) for 6 weeks beginning on postnatal day 28. SI and GH rats were tested in adulthood for anxiety-like behaviors (elevated plus maze), and the effects of EtOH (1 and 2 g/kg; intraperitoneally.) on NAc DA and NE were assessed by microdialysis. RESULTS: SI animals showed increased anxiety-like behavior compared to GH animals. Although SI had no effect on baseline levels of DA or NE, baseline DA levels were positively correlated with anxiety measures. In addition, while no significant differences were observed with 1 g/kg EtOH, the 2 g/kg dose induced significantly greater DA release in SI animals. Moreover, EtOH (2 g/kg) only elevated NAc NE levels in SI rats. CONCLUSIONS: These results suggest that chronic early-life stress sensitizes accumbal DA and NE release in response to an acute EtOH challenge. A greater EtOH sensitivity of DA and NE release dynamics in the NAc may contribute to increases in behavioral risk factors of alcoholism, like greater EtOH self-administration, that are observed in SI rats.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Dopamina/metabolismo , Etanol/administración & dosificación , Norepinefrina/metabolismo , Núcleo Accumbens/metabolismo , Aislamiento Social , Consumo de Bebidas Alcohólicas/psicología , Animales , Masculino , Núcleo Accumbens/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Long-Evans , Autoadministración , Aislamiento Social/psicología
10.
Alcohol Clin Exp Res ; 38(8): 2260-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25156616

RESUMEN

BACKGROUND: Methylphenidate (MPH) is a stimulant prescribed to treat attention-deficit/ hyperactivity disorder. Its primary mechanism of action is in the dopamine system, alterations of which are associated with vulnerability to alcohol abuse. There are concerns that juvenile MPH treatment may influence adult drinking behavior. This study examined the interaction of MPH treatment and environmental rearing conditions, which are known to independently influence ethanol (EtOH) drinking behavior, on anxiety-like behavior and vulnerability to alcohol abuse in a juvenile rodent model. METHODS: Male Sprague-Dawley rats were housed in enriched, standard, or isolated conditions for 4 weeks, starting at postnatal day 21. Rats were concurrently treated with 8 mg/kg/d MPH or saline, delivered via osmotic minipump. Anxiety-like behavior was determined at the end of the treatment session, and 5 weeks later. After MPH treatment, rats were exposed to a 2-bottle choice EtOH drinking procedure that lasted 3 weeks. RESULTS: Early life chronic MPH treatment was associated with greater EtOH intake and greater EtOH preference, but only in socially isolated animals. Isolated animals had greater levels of anxiety-like behavior than standard-housed or enriched animals after 4 weeks of exposure to the housing conditions, a difference that persisted even after all animals had been individually housed for an additional 5 weeks and exposed to EtOH. CONCLUSIONS: These results suggest that early life MPH treatment may increase vulnerability to EtOH drinking in adulthood in a subset of the population. Additionally, this study highlights the importance of early rearing condition for establishing long-lasting behavioral phenotypes. Environmental histories should be considered when prescribing MPH treatment to young children.


Asunto(s)
Consumo de Bebidas Alcohólicas/psicología , Metilfenidato/administración & dosificación , Metilfenidato/farmacología , Aislamiento Social/psicología , Factores de Edad , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Conducta de Elección/efectos de los fármacos , Masculino , Ratas
11.
Alcohol Clin Exp Res (Hoboken) ; 48(5): 903-917, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38472151

RESUMEN

BACKGROUND: Ethanol self-administration is governed by appetitive and consummatory behaviors. The sipper model procedurally separates these behaviors by training rats to meet a response requirement within 20 min to obtain continuous access to a sipper tube for an additional 20 min. Variations of this paradigm have been developed to quantify appetitive strength by evaluating lever presses during an extinction probe trial (EPT) or by deriving a break point (BP) from a progressive ratio (PR) schedule of reinforcement. However, no study has assessed the relationship between these tasks, within subjects, in both sexes. METHODS: Male and female rats (n = 16) were trained to meet a response requirement of 20 to access a slightly sweetened ethanol solution (10% ethanol + 1% sucrose). Two EPTs, during which no operant behavior was reinforced, were interleaved between 18 reinforced sessions. Next, rats completed an across-session PR schedule, where the response requirement increased each session. BP was defined as the highest completed response requirement. We then replicated the methodology in the same subjects responding for a 3% sucrose solution. Finally, the experiment was replicated in a separate cohort of rats (n = 24) trained to a response requirement of 4 to earn access to the ethanol solution and paradigm order (EPT vs. PR) was counterbalanced. RESULTS: We report strong, positive correlations between average EPT lever presses and BP across all experiments. No sex differences were observed in appetitive behaviors. However, the two cohorts revealed mixed results when assessing sex differences in consummatory measures. CONCLUSIONS: This study further validates the EPT as a measure of motivation and suggests that similar levels of motivation exist to procure alcohol in males and females. The findings complement the literature showing that appetitive and consummatory processes are distinct and thus should be independently assessed in self-administration paradigms.

12.
Eur J Neurosci ; 37(6): 1022-31, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23294165

RESUMEN

Social isolation (SI) rearing, a model of early life stress, results in profound behavioral alterations, including increased anxiety-like behavior, impaired sensorimotor gating and increased self-administration of addictive substances. These changes are accompanied by alterations in mesolimbic dopamine function, such as increased dopamine and metabolite tissue content, increased dopamine responses to cues and psychostimulants, and increased dopamine neuron burst firing. Using voltammetric techniques, we examined the effects of SI rearing on dopamine transporter activity, vesicular release and dopamine D2-type autoreceptor activity in the nucleus accumbens core. Long-Evans rats were housed in group (GH; 4/cage) or SI (1/cage) conditions from weaning into early adulthood [postnatal day (PD) 28-77]. After this initial housing period, rats were assessed on the elevated plus-maze for an anxiety-like phenotype, and then slice voltammetry experiments were performed. To study the enduring effects of SI rearing on anxiety-like behavior and dopamine terminal function, another cohort of similarly reared rats was isolated for an additional 4 months (until PD 174) and then tested. Our findings demonstrate that SI rearing results in lasting increases in anxiety-like behavior, dopamine release and dopamine transporter activity, but not D2 activity. Interestingly, GH-reared rats that were isolated as adults did not develop the anxiety-like behavior or dopamine changes seen in SI-reared rats. Together, our data suggest that early life stress results in an anxiety-like phenotype, with lasting increases in dopamine terminal function.


Asunto(s)
Ansiedad/metabolismo , Dopamina/metabolismo , Núcleo Accumbens/fisiología , Aislamiento Social , Factores de Edad , Animales , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Exocitosis , Núcleo Accumbens/crecimiento & desarrollo , Núcleo Accumbens/metabolismo , Ratas , Ratas Long-Evans , Receptores de Dopamina D2/metabolismo , Transducción de Señal
13.
J Exp Anal Behav ; 119(3): 488-500, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36788660

RESUMEN

The progression of recreational drinking to alcohol use disorder is characterized by loss of control over seeking, which involves continued use of alcohol despite negative consequences. The present study proposes a novel maladaptive alcohol self-administration task in which animals are trained to withhold alcohol drinking in the presence of an auditory cue signaling consequence (conflict phase) but to drink freely when there is no consequence (neutral phase). These phases are performed within trial; successful performance involves waiting for the conflict phase to end and drinking during the neutral phase. We discuss the background and implementation of the task, its relation to existing models, and its relevance to the field of translational alcohol research. Importantly, we also present evidence of its efficacy. Both male and female Long-Evans rats are capable of performing the maladaptive alcohol self-administration task for both sweetened and unsweetened alcohol solutions. Finally, we show that acute injection of a pharmacological stressor (yohimbine) significantly disrupted performance of the task in both sexes and reinforcers. We suggest the maladaptive alcohol self-administration task may prove particularly useful in models of alcohol use disorder or vulnerability to this disorder where its application may reveal maladaptive neural circuit adaptations responsible for motivational perturbations associated with loss of control over alcohol seeking.


Asunto(s)
Alcoholismo , Femenino , Masculino , Ratas , Animales , Ratas Long-Evans , Etanol/farmacología , Consumo de Bebidas Alcohólicas , Autoadministración , Condicionamiento Operante
14.
Addict Neurosci ; 92023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38188062

RESUMEN

Early life stress (ELS) is a major risk factor for alcohol use disorder (AUD) and comorbid neuropsychiatric conditions. We previously demonstrated that an adolescent social isolation (aSI) model of ELS significantly increased behavioral risk factors for these disorders (e.g. anxiety-like behaviors, alcohol drinking) in male, but not female rats. Since many neurodevelopmental milestones are accelerated in females, we investigated whether an earlier/shorter isolation window (PND 21-38) would yield comparable phenotypes in both sexes. In two experiments, Long Evans rats were socially isolated (SI) or group-housed (GH) on postnatal day (PND) 21 and locomotion was assessed in the open field test (OFT; PND 30). Experiment 1 also assessed behavior on the elevated plus-maze (EPM) (PND 32). In Experiment 2, all rats were single housed on PND 38 to assess home cage alcohol drinking. Experiment 1 revealed that SI females had increased locomotor activity in the OFT but did not differ from GH subjects on the EPM. The OFT results were replicated in both sexes in Experiment 2 and both male and female SI rats had significantly greater ethanol consumption during an eight day continuous access paradigm. In contrast, during subsequent intermittent two-bottle choice drinking, only SI females displayed greater ethanol intake and preference and increased consumption of a quinine-adulterated alcohol solution. These findings demonstrate that early life social isolation can promote AUD vulnerability-related phenotypes in female rats but that there are profound sex differences in the vulnerability window to this early life stressor. Uncovering the neural mechanisms responsible for these sexually dimorphic differences in sensitivity to ELS may shed light on the biological substrates associated with vulnerability to AUD and comorbid disorders of negative emotion in men and women.

15.
Sci Rep ; 11(1): 8749, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33888757

RESUMEN

The hippocampus, particularly its ventral domain, can promote negative affective states (i.e. stress and anxiety) that play an integral role in the development and persistence of alcohol use disorder (AUD). The ventral hippocampus (vHC) receives strong excitatory input from the basolateral amygdala (BLA) and the BLA-vHC projection bidirectionally modulates anxiety-like behaviors. However, no studies have examined the effects of chronic alcohol on the BLA-vHC circuit. In the present study, we used ex vivo electrophysiology in conjunction with optogenetic approaches to examine the effects of chronic intermittent ethanol exposure (CIE), a well-established rodent model of AUD, on the BLA-vHC projection and putative intrinsic vHC synaptic plasticity. We discovered prominent BLA innervation in the subicular region of the vHC (vSub). CIE led to an overall increase in the excitatory/inhibitory balance, an increase in AMPA/NMDA ratios but no change in paired-pulse ratios, consistent with a postsynaptic increase in excitability in the BLA-vSub circuit. CIE treatment also led to an increase in intrinsic network excitability in the vSub. Overall, our findings suggest a hyperexcitable state in BLA-vSub specific inputs as well as intrinsic inputs to vSub pyramidal neurons which may contribute to the negative affective behaviors associated with CIE.


Asunto(s)
Complejo Nuclear Basolateral/efectos de los fármacos , Etanol/farmacología , Hipocampo/efectos de los fármacos , Alcoholismo/fisiopatología , Animales , Complejo Nuclear Basolateral/fisiología , Etanol/administración & dosificación , Hipocampo/fisiología , Masculino , Plasticidad Neuronal/efectos de los fármacos , Optogenética , Ratas , Ratas Long-Evans , Transmisión Sináptica/efectos de los fármacos
16.
Alcohol ; 91: 41-51, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33321179

RESUMEN

Early life stress is known to impact vulnerability to psychopathological disorders in adulthood, including anxiety and alcohol use disorder (AUD), but the mechanisms underlying susceptibility to these outcomes are not fully understood. In the current study, we used adolescent social isolation (ASI) to determine whether Heterogeneous Stock (HS) rats, an outbred model used for genetic fine-mapping, could be used to study the genetics contributing to ASI-induced anxiety- and AUD-like behavior. We isolated (ASI) or group-housed (adolescent group-housed; AGH) 64 male HS rats at 4 weeks of age. After 5 weeks in these housing conditions, multiple anxiety and coping/despair-like behaviors were measured. All rats were then individually housed and assessed for voluntary ethanol self-administration. At euthanasia, synaptoneurosomes were isolated from a subset of brains to examine the expression of two proteins associated with alcohol drinking-related behaviors, GluA1 and SK2, in the dorsal (dHC) and ventral hippocampus (vHC). We found that ASI increased hyperactivity in the open field test relative to AGH, with no changes in other anxiety-like behaviors. Surprisingly, ASI rats demonstrated decreased immobility and increased climbing in the forced swim test relative to AGH. In contrast to prior studies by us and others, we found no difference in self-administration of 20% ethanol, with decreased ethanol self-administration in ASI relative to AGH rats at higher ethanol concentrations. Furthermore, while ASI in Long-Evans rats resulted in decreased SK2 expression in vHC synaptosomes, no differences were seen in vHC synaptosomes for SK2 or GluA1 in HS rats. These results demonstrate that HS rats are protected against many of the negative effects previously seen in response to ASI, namely anxiety-like behavior and increased ethanol self-administration. The current work suggests that a lack of change in SK2 and GluA1 expression levels in the vHC may play a role in conferring this protection.


Asunto(s)
Consumo de Bebidas Alcohólicas , Ansiedad , Hipercinesia/psicología , Estrés Psicológico/complicaciones , Animales , Etanol , Masculino , Ratas , Ratas Long-Evans
17.
Front Neurosci ; 15: 669075, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33994940

RESUMEN

Alcohol use disorder (AUD) differentially impacts men and women and a growing body of evidence points to sex-dependent adaptations in a number of brain regions. In a prior study, we explored the effect of a chronic intermittent ethanol exposure (CIE) model of AUD on neuronal and molecular adaptations in the dorsal and ventral domains of the hippocampus (dHC and vHC, respectively) in male rats. We found the vHC to be particularly sensitive to CIE, showing an increase in neuronal excitability and synaptic proteins associated with augmented excitation. These findings were accompanied by a CIE-dependent increase in anxiety-like behaviors. To explore sex-dependent adaptations in the hippocampus, we conducted a similar study in female rats. CIE-treated female rats showed a relatively modest increase in anxiety-like behaviors along with a robust increase in depressive-like measures. Despite both sexes showing clear evidence of a negative affective state following CIE, the vHC of females showed a decrease, rather than an increase, in neuronal excitability. In line with the reduced sensitivity to neural adaptations in the dHC of male rats, we were unable to identify any functional changes in the dHC of females. The functional changes of the vHC in female rats could not be explained by altered expression levels of a number of proteins typically associated with changes in neuronal excitability. Taken together, these findings point to sex as a major factor in CIE-dependent hippocampal adaptations that should be explored further to better understand possible gender differences in the etiology and treatment of AUD.

18.
Acta Neuropathol Commun ; 7(1): 173, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31703603

RESUMEN

Adolescent binge drinking represents a major public health challenge and can lead to persistent neurological and mental conditions, but the underlying pathogenic mechanisms remain poorly understood. Using a mouse model of adolescent binge ethanol treatment (ABET), we found that this treatment induced behavioral changes associated with demyelination in different brain regions. After ABET, adolescent mice exhibited anxiogenic behaviors with no change in locomotion on the elevated plus maze, and impaired spatial memory indicated by a significant reduction in spontaneous alternation in the Y maze test. Both effects persisted into adulthood. Anatomical studies further showed that ABET induced a significant reduction of parvalbumin-positive (PV+) GABAergic interneurons and myelin density in the hippocampus and medial prefrontal cortex (mPFC). While these deficits in PV+ interneurons and myelin persisted into early adulthood in the hippocampus, the myelin density recovered in the mPFC. Moreover, whereas ABET mainly damaged myelin of PV+ axons in the hippocampus, it primarily damaged myelin of PV-negative axons in the mPFC. Thus, our findings reveal that an adolescent binge alcohol treatment regimen disrupts spatial working memory, increases anxiety-like behaviors, and exerts unique temporal and spatial patterns of gray matter demyelination in the hippocampus and mPFC.


Asunto(s)
Ansiedad/patología , Consumo Excesivo de Bebidas Alcohólicas/patología , Enfermedades Desmielinizantes/patología , Etanol/toxicidad , Interneuronas/patología , Factores de Edad , Animales , Ansiedad/inducido químicamente , Ansiedad/psicología , Consumo Excesivo de Bebidas Alcohólicas/psicología , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/psicología , Etanol/administración & dosificación , Hipocampo/efectos de los fármacos , Hipocampo/patología , Interneuronas/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/patología
19.
Neuroscience ; 398: 144-157, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30481568

RESUMEN

Many studies have implicated hippocampal dysregulation in the pathophysiology of alcohol use disorder (AUD). However, over the past twenty years, a growing body of evidence has revealed distinct functional roles of the dorsal (dHC) and ventral (vHC) hippocampal subregions, with the dHC being primarily involved in spatial learning and memory and the vHC regulating anxiety- and depressive-like behaviors. Notably, to our knowledge, no rodent studies have examined the effects of chronic ethanol exposure on synaptic transmission along the dorsal/ventral axis. To that end, we examined the effects of the chronic intermittent ethanol vapor exposure (CIE) model of AUD on dHC and vHC synaptic excitability. Adult male Long-Evans rats were exposed to CIE or AIR for 10 days (12 h/day; targeting blood ethanol levels of 175-225 mg%) and recordings were made 24 h into withdrawal. As expected, this protocol increased anxiety-like behaviors on the elevated plus-maze and successive alleys test. Extracellular recordings revealed marked CIE-associated increases in synaptic excitation in the CA1 region that were exclusively restricted to the ventral domain of the hippocampus. Western blot analysis of synaptoneurosomal fractions revealed that the expression of two proteins that regulate synaptic strength, GluA2 and SK2, were dysregulated in the vHC, but not the dHC, following CIE. Together, these findings suggest that the ventral CA1 region may be particularly sensitive to the maladaptive effects of chronic ethanol exposure and provide new insight into some of the neural substrates that may contribute to the negative affective state that develops during withdrawal.


Asunto(s)
Trastornos Relacionados con Alcohol/fisiopatología , Depresores del Sistema Nervioso Central/efectos adversos , Etanol/efectos adversos , Hipocampo/efectos de los fármacos , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Quinasas del Centro Germinal , Hipocampo/fisiopatología , Masculino , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas Long-Evans , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/fisiología , Transmisión Sináptica/fisiología
20.
ACS Chem Neurosci ; 10(4): 2033-2044, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30284806

RESUMEN

Chronic peri-adolescent stress in humans increases risk to develop a substance use disorder during adulthood. Rats reared in social isolation during peri-adolescence (aSI; 1 rat/cage) period show greater ethanol and cocaine intake compared to group housed (aGH; 4 rats/cage) rats. In addition, aSI rats have a heightened dopamine response in the nucleus accumbens (NAc) to rewarding and aversive stimuli. Furthermore, single pulse electrical stimulation in slices containing NAc core elicits greater dopamine release in aSI rats. Here, we further investigated dopamine release kinetics and machinery following aSI. Dopamine release, across a wide range of stimulation intensities and frequencies, was significantly greater in aSI rats. Interestingly, subthreshold intensity stimulations also resulted in measurable dopamine release in accumbal slices from aSI but not aGH rats. Extracellular [Ca2+] manipulations revealed augmented calcium sensitivity of dopamine release in aSI rats. The readily releasable pools of dopamine, examined by bath application of Ro-04-1284/000, a vesicular monoamine transporter 2 (VMAT2) inhibitor, were depleted faster in aGH rats. Western blot analysis of release machinery proteins (VMAT2, Synaptogyrin-3, Syntaxin-1, and Munc13-3) showed no difference between the two groups. Tyrosine hydroxylase (TH) protein expression levels, however, were elevated in aSI rats. The greater dopamine release could potentially be explained by higher levels of TH, the rate-limiting step for dopamine synthesis. This augmented responsivity of the dopamine system and heightened dopamine availability post-aSI may lead to an increased risk of addiction vulnerability.


Asunto(s)
Dopamina/metabolismo , Núcleo Accumbens/metabolismo , Terminales Presinápticos/metabolismo , Aislamiento Social , Estrés Psicológico/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Factores de Edad , Animales , Enfermedad Crónica , Inhibidores de Captación de Dopamina/farmacología , Masculino , Núcleo Accumbens/efectos de los fármacos , Terminales Presinápticos/efectos de los fármacos , Ratas , Ratas Long-Evans , Aislamiento Social/psicología , Estrés Psicológico/psicología , Proteínas de Transporte Vesicular de Monoaminas/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA