Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
J Neurosci ; 43(50): 8690-8699, 2023 12 13.
Article in English | MEDLINE | ID: mdl-37932105

ABSTRACT

Avoidance stress coping, defined as persistent internal and/or external avoidance of stress-related stimuli, is a key feature of anxiety- and stress-related disorders, and contributes to increases in alcohol misuse after stress exposure. Previous work using a rat model of predator odor stress avoidance identified corticotropin-releasing factor (CRF) signaling via CRF Type 1 receptors (CRF1) in the CeA, as well as CeA projections to the lateral hypothalamus (LH) as key mediators of conditioned avoidance of stress-paired contexts and/or increased alcohol drinking after stress. Here, we report that CRF1-expressing CeA cells that project to the LH are preferentially activated in male and female rats that show persistent avoidance of predator odor stress-paired contexts (termed Avoider rats), and that chemogenetic inhibition of these cells rescues stress-induced increases in anxiety-like behavior and alcohol self-administration in male and female Avoider rats. Using slice electrophysiology, we found that prior predator odor stress exposure blunts inhibitory synaptic transmission and increases synaptic drive in CRF1 CeA-LH cells. In addition, we found that CRF bath application reduces synaptic drive in CRF1 CeA-LH cells in Non-Avoiders only. Collectively, these data show that CRF1 CeA-LH cells contribute to stress-induced increases in anxiety-like behavior and alcohol self-administration in male and female Avoider rats.SIGNIFICANCE STATEMENT Stress may lead to a variety of behavioral and physiological negative consequences, and better understanding of the neurobiological mechanisms that contribute to negative stress effects may lead to improved prevention and treatment strategies. This study, performed in laboratory rats, shows that animals that exhibit avoidance stress coping go on to develop heightened anxiety-like behavior and alcohol self-administration, and that these behaviors can be rescued by inhibiting the activity of a specific population of neurons in the central amygdala. This study also describes stress-induced physiological changes in these neurons that may contribute to their role in promoting increased anxiety and alcohol self-administration.


Subject(s)
Anxiety , Central Amygdaloid Nucleus , Corticotropin-Releasing Hormone , Ethanol , Stress Disorders, Traumatic , Animals , Female , Male , Rats , Anxiety/etiology , Central Amygdaloid Nucleus/metabolism , Corticotropin-Releasing Hormone/metabolism , Ethanol/administration & dosage , Hypothalamic Area, Lateral/metabolism , Neurons/physiology , Receptors, Corticotropin-Releasing Hormone/metabolism , Stress Disorders, Traumatic/complications
3.
Elife ; 112022 04 07.
Article in English | MEDLINE | ID: mdl-35389341

ABSTRACT

Corticotropin-releasing factor type-1 (CRF1) receptors are critical to stress responses because they allow neurons to respond to CRF released in response to stress. Our understanding of the role of CRF1-expressing neurons in CRF-mediated behaviors has been largely limited to mouse experiments due to the lack of genetic tools available to selectively visualize and manipulate CRF1+ cells in rats. Here, we describe the generation and validation of a transgenic CRF1-Cre-tdTomato rat. We report that Crhr1 and Cre mRNA expression are highly colocalized in both the central amygdala (CeA), composed of mostly GABAergic neurons, and in the basolateral amygdala (BLA), composed of mostly glutamatergic neurons. In the CeA, membrane properties, inhibitory synaptic transmission, and responses to CRF bath application in tdTomato+ neurons are similar to those previously reported in GFP+ cells in CRFR1-GFP mice. We show that stimulatory DREADD receptors can be targeted to CeA CRF1+ cells via virally delivered Cre-dependent transgenes, that transfected Cre/tdTomato+ cells are activated by clozapine-n-oxide in vitro and in vivo, and that activation of these cells in vivo increases anxiety-like and nocifensive behaviors. Outside the amygdala, we show that Cre-tdTomato is expressed in several brain areas across the brain, and that the expression pattern of Cre-tdTomato cells is similar to the known expression pattern of CRF1 cells. Given the accuracy of expression in the CRF1-Cre rat, modern genetic techniques used to investigate the anatomy, physiology, and behavioral function of CRF1+ neurons can now be performed in assays that require the use of rats as the model organism.


Subject(s)
Central Amygdaloid Nucleus , Corticotropin-Releasing Hormone , Animals , Anxiety , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Integrases , Mice , Nociception , Rats , Rats, Transgenic , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism
4.
J Neurosci ; 41(1): 61-72, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33188067

ABSTRACT

Persistent avoidance of stress-related stimuli following acute stress exposure predicts negative outcomes such as substance abuse and traumatic stress disorders. Previous work using a rat model showed that the central amygdala (CeA) plays an important role in avoidance of a predator odor stress-paired context. Here, we show that CeA projections to the lateral hypothalamus (LH) are preferentially activated in male rats that show avoidance of a predator odor-paired context (termed Avoider rats), that chemogenetic inhibition of CeA-LH projections attenuates avoidance in male Avoider rats, that chemogenetic stimulation of the CeA-LH circuit produces conditioned place avoidance (CPA) in otherwise naive male rats, and that avoidance behavior is associated with intrinsic properties of LH-projecting CeA cells. Collectively, these data show that CeA-LH projections are important for persistent avoidance of stress-related stimuli following acute stress exposure.SIGNIFICANCE STATEMENT This study in rats shows that a specific circuit in the brain [i.e., neurons that project from the central amygdala (CeA) to the lateral hypothalamus (LH)] mediates avoidance of stress-associated stimuli. In addition, this study shows that intrinsic physiological properties of cells in this brain circuit are associated with avoidance of stress-associated stimuli. Further characterization of the CeA-LH circuit may improve our understanding of the neural mechanisms underlying specific aspects of stress-related disorders in humans.


Subject(s)
Avoidance Learning/physiology , Central Amygdaloid Nucleus/physiology , Hypothalamic Area, Lateral/physiology , Neural Pathways/physiology , Animals , Behavior, Animal/physiology , Central Amygdaloid Nucleus/cytology , Electrophysiological Phenomena , Hypothalamic Area, Lateral/cytology , Male , Neural Pathways/cytology , Neurons , Odorants , Rats , Rats, Wistar
5.
Neuropharmacology ; 166: 107979, 2020 04.
Article in English | MEDLINE | ID: mdl-32028150

ABSTRACT

Post-traumatic stress disorder (PTSD) is characterized by avoidance of trauma-associated stimuli and amygdala hyperreactivity, and is highly co-morbid with alcohol use disorder (AUD). Our lab uses a predator odor (bobcat urine) stress model that produces conditioned avoidance of an odor-paired context in a subset of rats, mirroring avoidance symptoms that manifest in some but not all humans exposed to trauma. We previously showed that after predator odor stress, Avoiders exhibit escalated operant alcohol self-administration (SA), higher aversion-resistant operant alcohol responding, hyperalgesia, and greater anxiety-like behavior compared to unstressed Controls. We also showed previously that systemic antagonism of corticotropin-releasing factor-1 receptors (CRFR1) reduced escalation of operant alcohol SA in rats not indexed for avoidance, that corticotropin-releasing factor (CRF) infusions into the central amygdala (CeA) produced conditioned place avoidance in stress-naïve rats, and that intra-CeA infusion of a CRFR1 antagonist reduced hyperalgesia in Avoiders. Here, we show that avoidance behavior is persistent after repeated predator odor exposure. In addition, Avoiders showed lower weight gain than Controls after predator odor re-exposure. In the brain, higher avoidance was correlated with higher number of c-Fos + cells and CRF immunoreactivity in the CeA. Finally, we show that intra-CeA CRFR1 antagonism reversed post-stress escalation of alcohol SA and reduced avoidance behavior in Avoiders. Collectively, these findings suggest that elucidation of the mechanisms by which CRFR1-gated CeA circuits regulate avoidance behavior and alcohol SA may lead to better understanding of the neural mechanisms underlying co-morbid PTSD and AUD.


Subject(s)
Alcohol Drinking/metabolism , Avoidance Learning/physiology , Central Amygdaloid Nucleus/metabolism , Corticotropin-Releasing Hormone/metabolism , Odorants , Predatory Behavior/physiology , Stress, Psychological/metabolism , Alcohol Drinking/psychology , Animals , Lynx , Male , Rats , Rats, Wistar , Stress, Psychological/psychology
6.
Alcohol Res ; 40(1)2019 10 17.
Article in English | MEDLINE | ID: mdl-31649835

ABSTRACT

In this review, the effects of stress on alcohol drinking are discussed. The interactions between biological stress systems and alcohol drinking are examined, with a focus on the hypothalamic pituitary adrenal axis, corticotropin releasing factor, dynorphin, neuropeptide Y, and norepinephrine systems. Findings from animal models suggest that these biological stress systems may be useful targets for medications development for alcohol use disorder and co-occurring stress-related disorders in humans.


Subject(s)
Alcohol Drinking/epidemiology , Stress, Psychological/epidemiology , Alcohol Drinking/physiopathology , Animals , Comorbidity , Corticotropin-Releasing Hormone/physiology , Dynorphins/physiology , Humans , Hypothalamo-Hypophyseal System/physiopathology , Neuropeptide Y/physiology , Norepinephrine/physiology , Pituitary-Adrenal System/physiopathology , Stress, Psychological/physiopathology
7.
Genes Brain Behav ; 18(3): e12515, 2019 03.
Article in English | MEDLINE | ID: mdl-30129253

ABSTRACT

Common genetic factors may contribute to the high comorbidity between tobacco smoking and alcohol use disorder. Here, we assessed behavioral and biological effects of nicotine in replicate mouse lines selectively bred for high (HAP2/3) or low alcohol preference (LAP2/3). In Experiment 1, free-choice (FC) oral nicotine and quinine intake were assessed in HAP2/3 and LAP2/3 mice. Effects of nicotinic acetylcholine receptor blockade by mecamylamine on nicotine intake in HAP2 mice were also examined. In Experiment 2, HAP2/3 and LAP2/3 mice were tested for differences in sensitivity to nicotine-induced taste conditioning. In Experiment 3, the effects of a single nicotine injection on nucleus accumbens (NAc) and dorsal striatum monoamine levels in HAP2/3 and LAP2/3 mice were tested. In Experiment 1, HAP2/3 mice showed greater nicotine intake and intake ratio than LAP2/3 mice. There were no line differences in quinine intake. Mecamylamine reduced nicotine intake and intake ratio in HAP2 mice. In Experiment 2, HAP2/3 mice showed weaker nicotine-induced conditioned taste aversion (CTA) compared with LAP2/3 mice. In Experiment 3, nicotine treatment increased NAc dopamine turnover across both HAP2/3 and LAP2/3 mouse lines. These results show that there is a positive genetic correlation between oral alcohol intake (high alcohol intake/preference selection phenotype) and oral nicotine intake and a negative genetic correlation between oral alcohol intake and sensitivity to nicotine-induced CTA.


Subject(s)
Alcoholism/genetics , Genotype , Nicotine/pharmacology , Reinforcement, Psychology , Tobacco Smoking/genetics , Animals , Biogenic Monoamines/metabolism , Female , Male , Mecamylamine/pharmacology , Mice , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism
8.
Proc Natl Acad Sci U S A ; 115(49): 12525-12530, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30401738

ABSTRACT

Docosahexaenoic acid (DHA) is an omega-3 fatty acid that is highly abundant in the brain and confers protection against numerous neurological diseases, yet the fundamental mechanisms regulating the enrichment of DHA in the brain remain unknown. Here, we have discovered that a member of the long-chain acyl-CoA synthetase family, Acsl6, is required for the enrichment of DHA in the brain by generating an Acsl6-deficient mouse (Acsl6-/-). Acsl6 is highly enriched in the brain and lipid profiling of Acsl6-/- tissues reveals consistent reductions in DHA-containing lipids in tissues highly abundant with Acsl6. Acsl6-/- mice demonstrate motor impairments, altered glutamate metabolism, and increased astrogliosis and microglia activation. In response to a neuroinflammatory lipopolysaccharide injection, Acsl6-/- brains show similar increases in molecular and pathological indices of astrogliosis compared with controls. These data demonstrate that Acsl6 is a key mediator of neuroprotective DHA enrichment in the brain.


Subject(s)
Brain/enzymology , Coenzyme A Ligases/metabolism , Docosahexaenoic Acids/metabolism , Animals , Brain/metabolism , Coenzyme A Ligases/genetics , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Male , Mice , Mice, Knockout , Microglia , Motor Activity
9.
J Neurosci ; 38(36): 7761-7773, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30054393

ABSTRACT

Alcohol withdrawal symptoms contribute to excessive alcohol drinking and relapse in alcohol-dependent individuals. Among these symptoms, alcohol withdrawal promotes hyperalgesia, but the neurological underpinnings of this phenomenon are not known. Chronic alcohol exposure alters cell signaling in the central nucleus of the amygdala (CeA), and the CeA is implicated in mediating alcohol dependence-related behaviors. The CeA projects to the periaqueductal gray (PAG), a region critical for descending pain modulation, and may have a role in alcohol withdrawal hyperalgesia. Here, we tested the roles of (1) CeA projections to PAG, (2) CeA melanocortin signaling, and (3) PAG µ-opioid receptor signaling in mediating thermal nociception and alcohol withdrawal hyperalgesia in male Wistar rats. Our results demonstrate that alcohol dependence reduces GABAergic signaling from CeA terminals onto PAG neurons and alters the CeA melanocortin system, that CeA-PAG projections and CeA melanocortin signaling mediate alcohol withdrawal hyperalgesia, and that µ-opioid receptors in PAG filter CeA effects on thermal nociception.SIGNIFICANCE STATEMENT Hyperalgesia is commonly seen in individuals with alcohol use disorder during periods of withdrawal, but the neurological underpinnings behind this phenomenon are not completely understood. Here, we tested whether alcohol dependence exerts its influence on pain modulation via effects on the limbic system. Using behavioral, optogenetic, electrophysiological, and molecular biological approaches, we demonstrate that central nucleus of the amygdala (CeA) projections to periaqueductal gray mediate thermal hyperalgesia in alcohol-dependent and alcohol-naive rats. Using pharmacological approaches, we show that melanocortin receptor-4 signaling in CeA alters alcohol withdrawal hyperalgesia, but this effect is not mediated directly at synaptic inputs onto periaqueductal gray-projecting CeA neurons. Overall, our findings support a role for limbic influence over the descending pain pathway and identify a potential therapeutic target for treating hyperalgesia in individuals with alcohol use disorder .


Subject(s)
Alcoholism/physiopathology , Amygdala/physiopathology , Hyperalgesia/physiopathology , Nerve Net/physiopathology , Periaqueductal Gray/physiopathology , Alcoholism/metabolism , Amygdala/metabolism , Animals , Hot Temperature , Hyperalgesia/metabolism , Male , Nerve Net/metabolism , Neural Pathways/metabolism , Neural Pathways/physiopathology , Neurons/metabolism , Periaqueductal Gray/metabolism , Rats , Rats, Wistar , Receptor, Melanocortin, Type 4/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction/physiology
10.
Alcohol Clin Exp Res ; 42(2): 432-443, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29144544

ABSTRACT

BACKGROUND: Studies show that repeated nicotine use associates with high alcohol consumption in humans and that nicotine exposure sometimes increases alcohol consumption in animal models. However, the relative roles of genetic predisposition to high alcohol consumption, the alcohol drinking patterns, and the timing of nicotine exposure both with respect to alcohol drinking and developmental stage remain unclear. The studies here manipulated all these variables, using mice selectively bred for differences in free-choice (FC) alcohol consumption to elucidate the role of genetics and nicotine exposure in alcohol consumption behaviors. METHODS: In Experiments 1 and 2, we assessed the effects of repeated nicotine (0, 0.5, or 1.5 mg/kg) injections immediately before binge-like (drinking-in-the-dark; Experiment 1) or during FC alcohol access (Experiment 2) on these alcohol drinking behaviors (immediately after injections and during re-exposure to alcohol access 14 days later) in adult high- (HAP2) and low-alcohol-preferring (LAP2) female mice (co-exposure model). In Experiments 3 and 4, we assessed the effects of repeated nicotine (0, 0.5, or 1.5 mg/kg) injections 14 days prior to binge-like and FC alcohol access on these alcohol drinking behaviors in adolescent HAP2 and LAP2 female mice (Experiment 3) or adult HAP2 female mice (Experiment 4). RESULTS: In Experiment 1, we found that repeated nicotine (0.5 and 1.5 mg/kg) and alcohol co-exposure significantly increased binge-like drinking behavior in HAP2 but not LAP2 mice during the re-exposure phase after a 14-day abstinence period. In Experiment 2, 1.5 mg/kg nicotine injections significantly reduced FC alcohol intake and preference in the third hour postinjection in HAP2 but not LAP2 mice. No significant effects of nicotine treatment on binge-like or FC alcohol drinking were observed in Experiments 3 and 4. CONCLUSIONS: These results show that the temporal parameters of nicotine and alcohol exposure, pattern of alcohol access, and genetic predisposition for alcohol preference influence nicotine's effects on alcohol consumption. These findings in selectively bred mice suggest that humans with a genetic history of alcohol use disorders may be more vulnerable to develop nicotine and alcohol co-use disorders.


Subject(s)
Alcohol Drinking/genetics , Behavior, Animal/drug effects , Binge Drinking/genetics , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Animals , Female , Genetic Predisposition to Disease , Mice , Models, Animal , Selective Breeding
11.
PLoS One ; 12(7): e0182142, 2017.
Article in English | MEDLINE | ID: mdl-28759616

ABSTRACT

Nicotinic acetylcholine receptors containing α4 subunits (α4ß2* nAChRs) are critical for nicotinic cholinergic transmission and the addictive action of nicotine. To identify specific activities of these receptors in the adult mouse brain, we coupled targeted deletion of α4 nAChR subunits with behavioral and and electrophysiological measures of nicotine sensitivity. A viral-mediated Cre/lox approach allowed us to delete α4 from ventral midbrain (vMB) neurons. We used two behavioral assays commonly used to assess the motivational effects of drugs of abuse: home-cage oral self-administration, and place conditioning. Mice lacking α4 subunits in vMB consumed significantly more nicotine at the highest offered nicotine concentration (200 µg/mL) compared to control mice. Deletion of α4 subunits in vMB blocked nicotine-induced conditioned place preference (CPP) without affecting locomotor activity. Acetylcholine-evoked currents as well as nicotine-mediated increases in synaptic potentiation were reduced in mice lacking α4 in vMB. Immunostaining verified that α4 subunits were deleted from both dopamine and non-dopamine neurons in the ventral tegmental area (VTA). These results reveal that attenuation of α4* nAChR function in reward-related brain circuitry of adult animals may increase nicotine intake by enhancing the rewarding effects and/or reducing the aversive effects of nicotine.


Subject(s)
Nicotine/metabolism , Receptors, Nicotinic/metabolism , Reward , Ventral Tegmental Area/metabolism , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Drug-Seeking Behavior , Female , Gene Deletion , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Receptors, Nicotinic/genetics , Synaptic Potentials , Ventral Tegmental Area/physiology
12.
Alcohol ; 58: 127-137, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27908524

ABSTRACT

Post-traumatic stress disorder (PTSD) and alcohol-use disorders have a high rate of co-occurrence, possibly because they are regulated by common genes. In support of this idea, mice selectively bred for high (HAP) alcohol preference show greater fear potentiated startle (FPS), a model for fear-related disorders such as PTSD, compared to mice selectively bred for low (LAP) alcohol preference. This positive genetic correlation between alcohol preference and FPS behavior suggests that the two traits may be functionally related. This study examined the effects of fear conditioning on alcohol consumption and the effects of alcohol consumption on the expression of FPS in male and female HAP2 and LAP2 mice. In experiment 1, alcohol consumption (g/kg) under continuous-access conditions was monitored daily for 4 weeks following a single fear-conditioning or control treatment (foot shock and no shock). FPS was assessed three times (once at the end of the 4-week alcohol access period, once at 24 h after removal of alcohol, and once at 6-8 days after removal of alcohol), followed by two more weeks of alcohol access. Results showed no change in alcohol consumption, but alcohol-consuming, fear-conditioned, HAP2 males showed increased FPS at 24 h during the alcohol abstinence period compared to control groups. In experiment 2, alcohol consumption under limited-access conditions was monitored daily for 4 weeks. Fear-conditioning or control treatments occurred four times during the first 12 days and FPS testing occurred four times during the second 12 days of the 4-week alcohol consumption period. Results showed that fear conditioning increased alcohol intake in both HAP2 and LAP2 mice immediately following the first conditioning session. Fear-conditioned HAP2 but not LAP2 mice showed greater alcohol intake compared to control groups on drinking days that occurred between fear conditioning and FPS test sessions. FPS did not change as a function of alcohol consumption in either line. These results in mice help shed light on how a genetic propensity toward high alcohol consumption may be related to the risk for developing PTSD and co-morbid alcohol-use disorders in humans.


Subject(s)
Alcohol Drinking/genetics , Conditioning, Psychological/physiology , Ethanol/administration & dosage , Fear/physiology , Reflex, Startle/genetics , Acoustic Stimulation/methods , Alcohol Drinking/psychology , Animals , Conditioning, Psychological/drug effects , Fear/drug effects , Fear/psychology , Female , Male , Mice , Random Allocation , Reflex, Startle/drug effects , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...