1
|
Melkumyan M, Randall PA, Silberman Y. Central amygdala neuroimmune signaling in alcohol use disorder. ADDICTION NEUROSCIENCE 2025; 14:100194. [PMID: 40336623 PMCID: PMC12058212 DOI: 10.1016/j.addicn.2024.100194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Alcohol Use Disorder (AUD) is a prevalent and debilitating condition characterized by an inability to control alcohol consumption despite adverse consequences. Current treatments for AUD, including FDA-approved medications such as naltrexone and acamprosate, have limited efficacy and compliance, underscoring the need for novel therapeutic approaches. The central amygdala (CeA) plays a crucial role in the development and maintenance of AUD, particularly aspects associated with stress and binge behaviors. Recent research indicates neuroimmune signaling in the CeA is emerging as a key factor in this process. Chronic alcohol consumption disrupts neuroimmune signaling, leading to altered cytokine expression and activation of glial cells, including astrocytes and microglia. These changes contribute to the dysregulation of neural circuits involved in reward and stress, perpetuating alcohol-seeking behavior and relapse. This review delves into how chronic alcohol exposure affects neuroimmune signaling in the CeA, contributing to the pathophysiology of AUD. By focusing on the impact of cytokine expression and glial cell activation, this review aims to elucidate the mechanisms by which neuroinflammation in the CeA influences alcohol-related behaviors. By providing a comprehensive overview of the current state of research, this review identifies potential therapeutic targets for AUD. Understanding the complex interplay between neuroimmune signaling and alcohol-related behaviors may pave the way for more effective treatments and improved outcomes for individuals struggling with AUD.
Collapse
Affiliation(s)
- Mariam Melkumyan
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, United States
| | - Patrick A. Randall
- Department of Anesthesiology, Penn State College of Medicine, United States
- Department of Pharmacology, Penn State College of Medicine, United States
| | - Yuval Silberman
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, United States
| |
Collapse
|
2
|
Borgonetti V, Vozella V, Ware T, Cruz B, Bullard R, Cravatt BF, Galeotti N, Roberto M. Excessive alcohol intake produces persistent mechanical allodynia and dysregulates the endocannabinoid system in the lumbar dorsal root ganglia of genetically-selected Marchigian Sardinian alcohol-preferring rats. Pharmacol Res 2024; 209:107462. [PMID: 39396766 PMCID: PMC11834946 DOI: 10.1016/j.phrs.2024.107462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/20/2024] [Accepted: 09/15/2024] [Indexed: 10/15/2024]
Abstract
Epidemiological data indicate a strong association between alcohol use disorder (AUD) and neuropathic pain. Genetically-selected Marchigian Sardinian alcohol-preferring (msP) rats exhibit a high preference for alcohol compared with their background strain (Wistar rats), but their sensitivity to mechanical allodynia after chronic alcohol exposure is unknown. The present study compared the development of mechanical allodynia between "low, non-pathological drinker" Wistar rats and "high drinker" msP rats using the two-bottle choice (2BC) free-access procedure. Several studies reported the involvement of endocannabinoids (eCBs) in modulating mechanical allodynia, but there are no data on their role in alcohol-related allodynia. Thus, the present study assessed eCBs and their related lipid species in lumbar dorsal root ganglia (DRG) and correlated them with mechanical allodynia in our model. We found that male and female msP rats developed persistent mechanical allodynia during protracted abstinence from alcohol, presenting no sign of recovery, as opposed to Wistar rats. This effect directly correlated with their total alcohol intake. Notably, we found a correlation between lower lumbar DRG 2-arachidonoylglycerol (2-AG) levels and the development of higher mechanical allodynia during abstinence in msP rats of both sexes but not in Wistar rats. Moreover, alcohol-exposed and abstinent msP and Wistar females but not males exhibited significant alterations of thromboxane B2 and prostaglandin E2/prostaglandin D2 compared with naive rats. These findings demonstrate that DRG 2-AG metabolism is altered in msP rats during prolonged abstinence and represents a potentially interesting pharmacological target for the treatment of mechanical allodynia during alcohol abstinence.
Collapse
Affiliation(s)
- Vittoria Borgonetti
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA; Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Valentina Vozella
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Tim Ware
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Bryan Cruz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Ryan Bullard
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy.
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
3
|
Vlkolinsky R, Khom S, Vozella V, Bajo M, Roberto M. Withdrawal from chronic alcohol impairs the serotonin-mediated modulation of GABAergic transmission in the infralimbic cortex in male rats. Neurobiol Dis 2024; 199:106590. [PMID: 38996987 PMCID: PMC11412312 DOI: 10.1016/j.nbd.2024.106590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
The infralimbic cortex (IL) is part of the medial prefrontal cortex (mPFC), exerting top-down control over structures that are critically involved in the development of alcohol use disorder (AUD). Activity of the IL is tightly controlled by γ-aminobutyric acid (GABA) transmission, which is susceptible to chronic alcohol exposure and withdrawal. This inhibitory control is regulated by various neuromodulators, including 5-hydroxytryptamine (5-HT; serotonin). We used chronic intermittent ethanol vapor inhalation exposure, a model of AUD, in male Sprague-Dawley rats to induce alcohol dependence (Dep) followed by protracted withdrawal (WD; 2 weeks) and performed ex vivo electrophysiology using whole-cell patch clamp to study GABAergic transmission in layer V of IL pyramidal neurons. We found that WD increased frequencies of spontaneous inhibitory postsynaptic currents (sIPSCs), whereas miniature IPSCs (mIPSCs; recorded in the presence of tetrodotoxin) were unaffected by either Dep or WD. The application of 5-HT (50 μM) increased sIPSC frequencies and amplitudes in naive and Dep rats but reduced sIPSC frequencies in WD rats. Additionally, 5-HT2A receptor antagonist M100907 and 5-HT2C receptor antagonist SB242084 reduced basal GABA release in all groups to a similar extent. The blockage of either 5-HT2A or 5-HT2C receptors in WD rats restored the impaired response to 5-HT, which then resembled responses in naive rats. Our findings expand our understanding of synaptic inhibition in the IL in AUD, indicating that antagonism of 5-HT2A and 5-HT2C receptors may restore GABAergic control over IL pyramidal neurons. SIGNIFICANCE STATEMENT: Impairment in the serotonergic modulation of GABAergic inhibition in the medial prefrontal cortex contributes to alcohol use disorder (AUD). We used a well-established rat model of AUD and ex vivo whole-cell patch-clamp electrophysiology to characterize the serotonin modulation of GABAergic transmission in layer V infralimbic (IL) pyramidal neurons in ethanol-naive, ethanol-dependent (Dep), and ethanol-withdrawn (WD) male rats. We found increased basal inhibition following WD from chronic alcohol and altered serotonin modulation. Exogenous serotonin enhanced GABAergic transmission in naive and Dep rats but reduced it in WD rats. 5-HT2A and 5-HT2C receptor blockage in WD rats restored the typical serotonin-mediated enhancement of GABAergic inhibition. Our findings expand our understanding of synaptic inhibition in the infralimbic neurons in AUD.
Collapse
Affiliation(s)
- Roman Vlkolinsky
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Sophia Khom
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Pharmaceutical Sciences, University of Vienna, Vienna, 1090, Austria.
| | - Valentina Vozella
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
4
|
Patel RR, Gandhi P, Spencer K, Salem NA, Erikson CM, Borgonetti V, Vlkolinsky R, Rodriguez L, Nadav T, Bajo M, Roberts AJ, Dayne Mayfield R, Roberto M. Functional and morphological adaptation of medial prefrontal corticotropin releasing factor receptor 1-expressing neurons in male mice following chronic ethanol exposure. Neurobiol Stress 2024; 31:100657. [PMID: 38983690 PMCID: PMC11231756 DOI: 10.1016/j.ynstr.2024.100657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 07/11/2024] Open
Abstract
Chronic ethanol dependence and withdrawal activate corticotropin releasing factor (CRF)-containing GABAergic neurons in the medial prefrontal cortex (mPFC), which tightly regulate glutamatergic pyramidal neurons. Using male CRF1:GFP reporter mice, we recently reported that CRF1-expressing (mPFCCRF1+) neurons predominantly comprise mPFC prelimbic layer 2/3 pyramidal neurons, undergo profound adaptations following chronic ethanol exposure, and regulate anxiety and conditioned rewarding effects of ethanol. To explore the effects of acute and chronic ethanol exposure on glutamate transmission, the impact of chronic alcohol on spine density and morphology, as well as persistent changes in dendritic-related gene expression, we employed whole-cell patch-clamp electrophysiology, diOlistic labeling for dendritic spine analysis, and dendritic gene expression analysis to further characterize mPFCCRF1+ and mPFCCRF1- prelimbic layer 2/3 pyramidal neurons. We found increased glutamate release in mPFCCRF1+ neurons with ethanol dependence, which recovered following withdrawal. In contrast, we did not observe significant changes in glutamate transmission in neighboring mPFCCRF1- neurons. Acute application of 44 mM ethanol significantly reduced glutamate release onto mPFCCRF1+ neurons, which was observed across all treatment groups. However, this sensitivity to acute ethanol was only evident in mPFCCRF1- neurons during withdrawal. In line with alterations in glutamate transmission, we observed a decrease in total spine density in mPFCCRF1+ neurons during dependence, which recovered following withdrawal, while again no changes were observed in mPFCCRF- neurons. Given the observed decreases in mPFCCRF1+ stubby spines during withdrawal, we then identified persistent changes at the dendritic gene expression level in mPFCCRF1+ neurons following withdrawal that may underlie these structural adaptations. Together, these findings highlight the varying responses of mPFCCRF1+ and mPFCCRF1- cell-types to acute and chronic ethanol exposure, as well as withdrawal, revealing specific functional, morphological, and molecular adaptations that may underlie vulnerability to ethanol and the lasting effects of ethanol dependence.
Collapse
Affiliation(s)
- Reesha R. Patel
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Pauravi Gandhi
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Kathryn Spencer
- Core Microscopy Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Nihal A. Salem
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Chloe. M. Erikson
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Vittoria Borgonetti
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Roman Vlkolinsky
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Larry Rodriguez
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Tali Nadav
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Amanda J. Roberts
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - R. Dayne Mayfield
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| |
Collapse
|
5
|
Melkumyan M, Annaswamy VM, Evans AM, Showemimo OF, McCullers ZE, Sun D, Murphy TE, Vrana KE, Arnold AC, Raup-Konsavage WM, Silberman Y. Effects of cannabidiol, with and without ∆9-tetrahydrocannabinol, on anxiety-like behavior following alcohol withdrawal in mice. Front Neurosci 2024; 18:1375440. [PMID: 38957186 PMCID: PMC11217543 DOI: 10.3389/fnins.2024.1375440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/29/2024] [Indexed: 07/04/2024] Open
Abstract
Introduction Alcohol use disorder (AUD) is commonly associated with anxiety disorders and enhanced stress-sensitivity; symptoms that can worsen during withdrawal to perpetuate continued alcohol use. Alcohol increases neuroimmune activity in the brain. Our recent evidence indicates that alcohol directly modulates neuroimmune function in the central amygdala (CeA), a key brain region regulating anxiety and alcohol intake, to alter neurotransmitter signaling. We hypothesized that cannabinoids, such as cannabidiol (CBD) and ∆9-tetrahydrocannabinol (THC), which are thought to reduce neuroinflammation and anxiety, may have potential utility to alleviate alcohol withdrawal-induced stress-sensitivity and anxiety-like behaviors via modulation of CeA neuroimmune function. Methods We tested the effects of CBD and CBD:THC (3:1 ratio) on anxiety-like behaviors and neuroimmune function in the CeA of mice undergoing acute (4-h) and short-term (24-h) withdrawal from chronic intermittent alcohol vapor exposure (CIE). We further examined the impact of CBD and CBD:THC on alcohol withdrawal behaviors in the presence of an additional stressor. Results We found that CBD and 3:1 CBD:THC increased anxiety-like behaviors at 4-h withdrawal. At 24-h withdrawal, CBD alone reduced anxiety-like behaviors while CBD:THC had mixed effects, showing increased center time indicating reduced anxiety-like behaviors, but increased immobility time that may indicate increased anxiety-like behaviors. These mixed effects may be due to altered metabolism of CBD and THC during alcohol withdrawal. Immunohistochemical analysis showed decreased S100β and Iba1 cell counts in the CeA at 4-h withdrawal, but not at 24-h withdrawal, with CBD and CBD:THC reversing alcohol withdrawal effects.. Discussion These results suggest that the use of cannabinoids during alcohol withdrawal may lead to exacerbated anxiety depending on timing of use, which may be related to neuroimmune cell function in the CeA.
Collapse
Affiliation(s)
- Mariam Melkumyan
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, Hershey, PA, United States
| | - Vibha M. Annaswamy
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, Hershey, PA, United States
| | - Alexandra M. Evans
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, Hershey, PA, United States
| | - Opeyemi F. Showemimo
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, Hershey, PA, United States
| | - Zari E. McCullers
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, Hershey, PA, United States
| | - Dongxiao Sun
- The Pennsylvania State University College of Medicine, Department of Pharmacology, Hershey, PA, United States
| | - Terrence E. Murphy
- The Pennsylvania State University College of Medicine, Department of Public Health Sciences, Hershey, PA, United States
| | - Kent E. Vrana
- The Pennsylvania State University College of Medicine, Department of Pharmacology, Hershey, PA, United States
| | - Amy C. Arnold
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, Hershey, PA, United States
| | - Wesley M. Raup-Konsavage
- The Pennsylvania State University College of Medicine, Department of Pharmacology, Hershey, PA, United States
| | - Yuval Silberman
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, Hershey, PA, United States
| |
Collapse
|
6
|
Foo JC, Skorodumov I, Spanagel R, Meinhardt MW. Sex- and age-specific effects on the development of addiction and compulsive-like drinking in rats. Biol Sex Differ 2023; 14:44. [PMID: 37420305 PMCID: PMC10327342 DOI: 10.1186/s13293-023-00529-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 06/27/2023] [Indexed: 07/09/2023] Open
Abstract
BACKGROUND Biological factors are known to influence disease trajectories and treatment effectiveness in alcohol addiction and preclinical and clinical evidence suggests that sex is an important factor influencing disease dynamics in alcohol dependence. Another critical factor is age at first intoxicating drink, which has been identified as a risk factor for later alcohol binging. Preclinical research allows prospective monitoring of rodents throughout the lifespan, providing very detailed information that cannot be acquired in humans. Lifetime monitoring in rodents can be conducted under highly controlled conditions, during which one can systematically introduce multiple biological and environmental factors that impact behaviors of interest. METHODS Here, we used the alcohol deprivation effect (ADE) rat model of alcohol addiction in a computerized drinkometer system, acquiring high-resolution data to study changes over the course of addictive behavior as well as compulsive-like drinking in cohorts of adolescent vs. adult as well as male vs. female rats. RESULTS Female rats drank more alcohol than male rats during the whole experiment, drinking much more weak alcohol (5%) and similar amounts of stronger alcohol solutions (10%, 20%); female rats also consumed more alcohol than male rats during quinine taste adulteration. Increased consumption in females compared to males was driven by larger access sizes of alcohol. Differences in circadian patterns of movement were observed between groups. Early age of onset of drinking (postnatal day 40) in male rats had surprisingly little impact on the development of drinking behavior and compulsivity (quinine taste adulteration) when compared to rats that started drinking during early adulthood (postnatal day 72). CONCLUSIONS Our results suggest that there are sex-specific drinking patterns, not only in terms of total amount consumed, but specifically in terms of solution preference and access size. These findings provide a better understanding of sex and age factors involved in the development of drinking behavior, and can inform the preclinical development of models of addiction, drug development and exploration of options for new treatments.
Collapse
Affiliation(s)
- Jerome C. Foo
- Department of Genetic Epidemiology in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
- Institute for Psychopharmacology, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Ivan Skorodumov
- Institute for Psychopharmacology, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Rainer Spanagel
- Institute for Psychopharmacology, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Marcus W. Meinhardt
- Institute for Psychopharmacology, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
- Department of Molecular Neuroimaging, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
7
|
Abstract
Ethanol (EtOH) has effects on numerous cellular molecular targets, and alterations in synaptic function are prominent among these effects. Acute exposure to EtOH activates or inhibits the function of proteins involved in synaptic transmission, while chronic exposure often produces opposing and/or compensatory/homeostatic effects on the expression, localization, and function of these proteins. Interactions between different neurotransmitters (e.g., neuropeptide effects on release of small molecule transmitters) can also influence both acute and chronic EtOH actions. Studies in intact animals indicate that the proteins affected by EtOH also play roles in the neural actions of the drug, including acute intoxication, tolerance, dependence, and the seeking and drinking of EtOH. The present chapter is an update of our previous Lovinger and Roberto (Curr Top Behav Neurosci 13:31-86, 2013) chapter and reviews the literature describing these acute and chronic synaptic effects of EtOH with a focus on adult animals and their relevance for synaptic transmission, plasticity, and behavior.
Collapse
Affiliation(s)
- David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism (NIAAA), Rockville, MD, USA
| | - Marisa Roberto
- Molecular Medicine Department, Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
8
|
Kim HJJ, Zagzoog A, Black T, Baccetto SL, Laprairie RB. Molecular and cellular mechanisms underlying brain region-specific endocannabinoid system modulation by estradiol across the rodent estrus cycle. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 195:27-45. [PMID: 36707154 DOI: 10.1016/bs.pmbts.2022.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Neurological crosstalk between the endocannabinoid and estrogen systems has been a growing topic of discussion over the last decade. Although the main estrogenic ligand, estradiol (E2), influences endocannabinoid signaling in both male and female animals, the latter experiences significant and rhythmic fluctuations in E2 as well as other sex hormones. This is referred to as the menstrual cycle in women and the estrus cycle in rodents such as mice and rats. Consisting of 4 distinct hormone-driven phases, the rodent estrus cycle modulates both endocannabinoid and exogenous cannabinoid signaling resulting in unique behavioral outcomes based on the cycle phase. For example, cannabinoid receptor agonist-induced antinociception is greatest during proestrus and estrus, when circulating and brain levels of E2 are high, as compared to metestrus and diestrus when E2 concentrations are low. Pain processing occurs throughout the cerebral cortex and amygdala of the forebrain; periaqueductal grey of the midbrain; and medulla and spine of the hindbrain. As a result, past molecular investigations on these endocannabinoid-estrogen system interactions have focused on these specific brain regions. Here, we will bridge regional molecular trends with neurophysiological evidence of how plasma membrane estrogen receptor (ER) activation by E2 leads to postsynaptic endocannabinoid synthesis, retrograde signaling, and alterations in inhibitory neurotransmission. These signaling pathways depend on ER heterodimers, current knowledge of which will also be detailed in this review. Overall, the aim of this review article is to systematically summarize how the cannabinoid receptors and endocannabinoids change in expression and function in specific brain regions throughout the estrus cycle.
Collapse
Affiliation(s)
- Hye Ji J Kim
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ayat Zagzoog
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Tallan Black
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Sarah L Baccetto
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Robert B Laprairie
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada; Department of Pharmacology, College of Medicine, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
9
|
Melkumyan M, Silberman Y. Subregional Differences in Alcohol Modulation of Central Amygdala Neurocircuitry. Front Mol Neurosci 2022; 15:888345. [PMID: 35866156 PMCID: PMC9294740 DOI: 10.3389/fnmol.2022.888345] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Alcohol use disorder is a highly significant medical condition characterized by an impaired ability to stop or control alcohol use, compulsive alcohol seeking behavior, and withdrawal symptoms in the absence of alcohol. Understanding how alcohol modulates neurocircuitry critical for long term and binge-like alcohol use, such as the central amygdala (CeA), may lead to the development of novel therapeutic strategies to treat alcohol use disorder. In clinical studies, reduction in the volume of the amygdala has been linked with susceptibility to relapse to alcohol use. Preclinical studies have shown the involvement of the CeA in the effects of alcohol use, with lesions of the amygdala showing a reduction in alcohol drinking, and manipulations of cells in the CeA altering alcohol drinking. A great deal of work has shown that acute alcohol, as well as chronic alcohol exposure via intake or dependence models, alters glutamatergic and GABAergic transmission in the CeA. The CeA, however, contains heterogeneous cell populations and distinct subregional differences in neurocircuit architecture which may influence the mechanism by which alcohol modulates CeA function overall. The current review aimed to parse out the differences in alcohol effects on the medial and lateral subregions of the CeA, and what role neuroinflammatory cells and markers, the endocannabinoid system, and the most commonly studied neuropeptide systems play in mediating these effects. A better understanding of alcohol effects on CeA subregional cell type and neurocircuit function may lead to development of more selective pharmacological interventions for alcohol use disorder.
Collapse
Affiliation(s)
- Mariam Melkumyan
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
| | - Yuval Silberman
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
10
|
Abstract
PURPOSE A growing body of evidence has implicated the endocannabinoid (eCB) system in the acute, chronic, and withdrawal effects of alcohol/ethanol on synaptic function. These eCB-mediated synaptic effects may contribute to the development of alcohol use disorder (AUD). Alcohol exposure causes neurobiological alterations similar to those elicited by chronic cannabinoid (CB) exposure. Like alcohol, cannabinoids alter many central processes, such as cognition, locomotion, synaptic transmission, and neurotransmitter release. There is a strong need to elucidate the effects of ethanol on the eCB system in different brain regions to understand the role of eCB signaling in AUD. SEARCH METHODS For the scope of this review, preclinical studies were identified through queries of the PubMed database. SEARCH RESULTS This search yielded 459 articles. Clinical studies and papers irrelevant to the topic of this review were excluded. DISCUSSION AND CONCLUSIONS The endocannabinoid system includes, but is not limited to, cannabinoid receptors 1 (CB1), among the most abundantly expressed neuronal receptors in the brain; cannabinoid receptors 2 (CB2); and endogenously formed CB1 ligands, including arachidonoylethanolamide (AEA; anandamide), and 2-arachidonoylglycerol (2-AG). The development of specific CB1 agonists, such as WIN 55,212-2 (WIN), and antagonists, such as SR 141716A (rimonabant), provide powerful pharmacological tools for eCB research. Alcohol exposure has brain region-specific effects on the eCB system, including altering the synthesis of endocannabinoids (e.g., AEA, 2-AG), the synthesis of their precursors, and the density and coupling efficacy of CB1. These alcohol-induced alterations of the eCB system have subsequent effects on synaptic function including neuronal excitability and postsynaptic conductance. This review will provide a comprehensive evaluation of the current literature on the synaptic interactions of alcohol exposure and eCB signaling systems, with an emphasis on molecular and physiological synaptic effects of alcohol on the eCB system. A limited volume of studies has focused on the underlying interactions of alcohol and the eCB system at the synaptic level in the brain. Thus, the data on synaptic interactions are sparse, and future research addressing these interactions is much needed.
Collapse
Affiliation(s)
- Sarah A Wolfe
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, California
| | - Valentina Vozella
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, California
| | - Marisa Roberto
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, California
| |
Collapse
|
11
|
Hitzemann R, Bergeson SE, Berman AE, Bubier JA, Chesler EJ, Finn DA, Hein M, Hoffman P, Holmes A, Kisby BR, Lockwood D, Lodowski KH, McManus M, Owen JA, Ozburn AR, Panthagani P, Ponomarev I, Saba L, Tabakoff B, Walchale A, Williams RW, Phillips TJ. Sex Differences in the Brain Transcriptome Related to Alcohol Effects and Alcohol Use Disorder. Biol Psychiatry 2022; 91:43-52. [PMID: 34274109 PMCID: PMC8558111 DOI: 10.1016/j.biopsych.2021.04.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/05/2021] [Accepted: 04/26/2021] [Indexed: 01/03/2023]
Abstract
There is compelling evidence that sex and gender have crucial roles in excessive alcohol (ethanol) consumption. Here, we review some of the data from the perspective of brain transcriptional differences between males and females, focusing on rodent animal models. A key emerging transcriptional feature is the role of neuroimmune processes. Microglia are the resident neuroimmune cells in the brain and exhibit substantial functional differences between males and females. Selective breeding for binge ethanol consumption and the impacts of chronic ethanol consumption and withdrawal from chronic ethanol exposure all demonstrate sex-dependent neuroimmune signatures. A focus is on resolving sex-dependent differences in transcriptional responses to ethanol at the neurocircuitry level. Sex-dependent transcriptional differences are found in the extended amygdala and the nucleus accumbens. Telescoping of ethanol consumption is found in some, but not all, studies to be more prevalent in females. Recent transcriptional studies suggest that some sex differences may be due to female-dependent remodeling of the primary cilium. An interesting theme appears to be developing: at least from the animal model perspective, even when males and females are phenotypically similar, they differ significantly at the level of the transcriptome.
Collapse
Affiliation(s)
- Robert Hitzemann
- Department of Behavioral Neuroscience and Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon
| | - Susan E Bergeson
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| | | | | | | | - Deborah A Finn
- Department of Behavioral Neuroscience and Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon; Veterans Affairs Portland Health Care System, Portland, Oregon
| | - Matthew Hein
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Paula Hoffman
- Department of Pharmacology, University of Colorado, Aurora, Colorado
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Brent R Kisby
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Denesa Lockwood
- Department of Behavioral Neuroscience and Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon.
| | - Kerrie H Lodowski
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Michelle McManus
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Julie A Owen
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Angela R Ozburn
- Department of Behavioral Neuroscience and Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon; Veterans Affairs Portland Health Care System, Portland, Oregon
| | - Praneetha Panthagani
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Igor Ponomarev
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Laura Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Boris Tabakoff
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Aashlesha Walchale
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Sciences Center, Memphis, Tennessee
| | - Tamara J Phillips
- Department of Behavioral Neuroscience and Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon; Veterans Affairs Portland Health Care System, Portland, Oregon
| |
Collapse
|
12
|
Rangel-Barajas C, Boehm SL, Logrip ML. Altered excitatory transmission in striatal neurons after chronic ethanol consumption in selectively bred crossed high alcohol-preferring mice. Neuropharmacology 2021; 190:108564. [PMID: 33857521 PMCID: PMC8293703 DOI: 10.1016/j.neuropharm.2021.108564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/24/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
Genetic predisposition to heavy drinking is a risk factor for alcohol misuse. We used selectively bred crossed high alcohol-preferring (cHAP) mice to study sex differences in alcohol drinking and its effect on glutamatergic activity in dorsolateral (DLS) and dorsomedial (DMS) striatum. We performed whole-cell patch-clamp recording in neurons from male and female cHAP mice with 5-week alcohol drinking history and alcohol-naïve controls. In DMS, alcohol-naïve males' neurons displayed lower cell capacitance and higher membrane resistance than females' neurons, both effects reversed by drinking. Conversely, in DLS neurons, drinking history increased capacitance only in males and changed membrane resistance only in females. Altered biophysical membrane properties were accompanied by disrupted glutamatergic transmission. Drinking history increased spontaneous excitatory postsynaptic current (sEPSC) amplitude in DMS and frequency in DLS female neurons, compared to alcohol-naïve females, without effect in males. Acute ethanol differentially impacted DMS and DLS neurons by sex and drinking history. In DMS, acute alcohol significantly increased sEPSC frequency only in neurons from alcohol-naïve females, an effect that disappeared after drinking history. In DLS, acute alcohol had opposing effects in males and females based on drinking history. Estrous cycle also impacted DMS and DLS neurons differently: sEPSC amplitudes were higher in DMS cells from drinking history than alcohol-naïve females, whereas estrous cycle, not drinking history, modified DLS firing rate. Our data show sex differences in cHAP ethanol consumption and neurophysiology, suggesting differential dysregulation of glutamatergic drive onto DMS and DLS after chronic ethanol consumption.
Collapse
Affiliation(s)
- Claudia Rangel-Barajas
- Department of Psychology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Stephen L Boehm
- Department of Psychology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, 46202, USA; Indiana Alcohol Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Marian L Logrip
- Department of Psychology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, 46202, USA; Indiana Alcohol Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
13
|
Natividad LA, Steinman MQ, McGinn MA, Sureshchandra S, Kerr TM, Ciccocioppo R, Messaoudi I, Edwards S, Roberto M. Impaired hypothalamic feedback dysregulates brain glucocorticoid signaling in genetically-selected Marchigian Sardinian alcohol-preferring rats. Addict Biol 2021; 26:e12978. [PMID: 33142367 PMCID: PMC8052265 DOI: 10.1111/adb.12978] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/17/2020] [Accepted: 09/30/2020] [Indexed: 12/20/2022]
Abstract
Genetically-selected Marchigian Sardinian alcohol-preferring (msP) rats display comorbid symptoms of increased alcohol preference and elevated anxiety-like behavior. Heightened stress sensitivity in msPs is influenced by genetic polymorphisms of the corticotropin-releasing factor receptor in the central nucleus of the amygdala (CeA), as well as reduced influence of anti-stress mechanisms that normally constrain the stress response. Given this propensity for stress dysregulation, in this study, we expand on the possibility that msPs may display differences in neuroendocrine processes that normally terminate the stress response. We utilized behavioral, biochemical, and molecular assays to compare basal and restraint stress-induced changes in the hypothalamic-pituitary-adrenal (HPA) axis of male and female msPs relative to their nonselected Wistar counterparts. The results showed that msPs display deficits in marble-burying behavior influenced by environmental factors and procedures that modulate arousal states in a sex-dependent manner. Whereas male msPs display evidence of dysregulated neuroendocrine function (higher adrenocorticotropic hormone levels and subthreshold reductions in corticosterone), females display restraint-induced elevations in corticosterone levels that were persistently higher in msPs. A dexamethasone challenge reduced the circulation of these stress hormones, although the reduction in corticosterone was generally attenuated in msP versus Wistar rats. Finally, we found evidence of diminished stress-induced glucocorticoid receptor (GR) phosphorylation in the hypothalamic paraventricular nucleus of msPs, as well as innate increases in phosphorylated GR levels in the CeA of male msPs. Collectively, these findings suggest that negative feedback processes regulating HPA responsiveness are diminished in msP rats, possibly underlying differences in the expression of anxiety-like behaviors.
Collapse
Affiliation(s)
- Luis A. Natividad
- College of Pharmacy, The University of Texas at Austin, Austin, Texas, 78712, USA
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, 92037, USA
| | - Michael Q. Steinman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, 92037, USA
| | - M. Adrienne McGinn
- Department of Physiology, Louisiana State University Health Science Center, New Orleans, Louisiana, 70112, USA
| | - Suhas Sureshchandra
- School of Biological Sciences, University of California at Irvine, Irvine, California, 92697, USA
| | - Tony M. Kerr
- College of Pharmacy, The University of Texas at Austin, Austin, Texas, 78712, USA
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, 92037, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Ilhem Messaoudi
- School of Biological Sciences, University of California at Irvine, Irvine, California, 92697, USA
| | - Scott Edwards
- Department of Physiology, Louisiana State University Health Science Center, New Orleans, Louisiana, 70112, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, 92037, USA
| |
Collapse
|
14
|
Kirson D, Khom S, Rodriguez L, Wolfe SA, Varodayan FP, Gandhi PJ, Patel RR, Vlkolinsky R, Bajo M, Roberto M. Sex Differences in Acute Alcohol Sensitivity of Naïve and Alcohol Dependent Central Amygdala GABA Synapses. Alcohol Alcohol 2021; 56:581-588. [PMID: 33912894 DOI: 10.1093/alcalc/agab034] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/26/2021] [Accepted: 04/10/2021] [Indexed: 01/08/2023] Open
Abstract
AIMS Alcohol use disorder (AUD) is linked to hyperactivity of brain stress systems, leading to withdrawal states which drive relapse. AUD differs among the sexes, as men are more likely to have AUD than women, but women progress from casual use to binge and heavy alcohol use more quickly and are more likely to relapse into repetitive episodes of heavy drinking. In alcohol dependence animal models of AUD, the central amygdala (CeA) functions as a hub of stress and anxiety processing and gamma-Aminobutyric acid (GABA)ergic signaling within the CeA is involved in dependence-induced increases in alcohol consumption. We have shown dysregulation of CeA GABAergic synaptic signaling in alcohol dependence animal models, but previous studies have exclusively used males. METHODS Here, we used whole-cell patch clamp electrophysiology to examine basal CeA GABAergic spontaneous inhibitory postsynaptic currents (sIPSC) and the effects of acute alcohol in both naïve and alcohol dependent rats of both sexes. RESULTS We found that sIPSC kinetics differ between females and males, as well as between naïve and alcohol-dependent animals, with naïve females having the fastest current kinetics. Additionally, we find differences in baseline current kinetics across estrous cycle stages. In contrast to the increase in sIPSC frequency routinely found in males, acute alcohol (11-88 mM) had no effect on sIPSCs in naïve females, however the highest concentration of alcohol increased sIPSC frequency in dependent females. CONCLUSION These results provide important insight into sex differences in CeA neuronal function and dysregulation with alcohol dependence and highlight the need for sex-specific considerations in the development of effective AUD treatment.
Collapse
Affiliation(s)
- Dean Kirson
- The Scripps Research Institute, Department of Molecular Medicine, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Sophia Khom
- The Scripps Research Institute, Department of Molecular Medicine, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Larry Rodriguez
- The Scripps Research Institute, Department of Molecular Medicine, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Sarah A Wolfe
- The Scripps Research Institute, Department of Molecular Medicine, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Florence P Varodayan
- The Scripps Research Institute, Department of Molecular Medicine, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Pauravi J Gandhi
- The Scripps Research Institute, Department of Molecular Medicine, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Reesha R Patel
- The Scripps Research Institute, Department of Molecular Medicine, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Roman Vlkolinsky
- The Scripps Research Institute, Department of Molecular Medicine, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Michal Bajo
- The Scripps Research Institute, Department of Molecular Medicine, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Marisa Roberto
- The Scripps Research Institute, Department of Molecular Medicine, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
15
|
Vozella V, Cruz B, Natividad LA, Benvenuti F, Cannella N, Edwards S, Zorrilla EP, Ciccocioppo R, Roberto M. Glucocorticoid Receptor Antagonist Mifepristone Does Not Alter Innate Anxiety-Like Behavior in Genetically-Selected Marchigian Sardinian (msP) Rats. Int J Mol Sci 2021; 22:3095. [PMID: 33803557 PMCID: PMC8003048 DOI: 10.3390/ijms22063095] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 12/17/2022] Open
Abstract
Marchigian Sardinian alcohol-preferring (msP) rats serve as a unique model of heightened alcohol preference and anxiety disorders. Their innate enhanced stress and poor stress-coping strategies are driven by a genetic polymorphism of the corticotropin-releasing factor receptor 1 (CRF1) in brain areas involved in glucocorticoid signaling. The activation of glucocorticoid receptors (GRs) regulates the stress response, making GRs a candidate target to treat stress and anxiety. Here, we examined whether mifepristone, a GR antagonist known to reduce alcohol drinking in dependent rats, decreases innate symptoms of anxiety in msPs. Male and female msPs were compared to non-selected Wistar counterparts across three separate behavioral tests. We assessed anxiety-like behavior via the novelty-induced hypophagia (NIH) assay. Since sleep disturbances and hyperarousal are common features of stress-related disorders, we measured sleeping patterns using the comprehensive lab monitoring system (CLAMS) and stress sensitivity using acoustic startle measures. Rats received an acute administration of vehicle or mifepristone (60 mg/kg) 90 min prior to testing on NIH, acoustic startle response, and CLAMS. Our results revealed that both male and female msPs display greater anxiety-like behaviors as well as enhanced acoustic startle responses compared to Wistar counterparts. Male msPs also displayed reduced sleeping bout duration versus Wistars, and female msPs displayed greater acoustic startle responses versus male msPs. Importantly, the enhanced anxiety-like behavior and startle responses were not reduced by mifepristone. Together, these findings suggest that increased expression of stress-related behaviors in msPs are not solely mediated by acute activation of GRs.
Collapse
Affiliation(s)
- Valentina Vozella
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (V.V.); (B.C.); (E.P.Z.)
| | - Bryan Cruz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (V.V.); (B.C.); (E.P.Z.)
| | - Luis A. Natividad
- College of Pharmacy, The University of Austin, Austin, TX 78712, USA;
| | - Federica Benvenuti
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy; (F.B.); (N.C.); (R.C.)
| | - Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy; (F.B.); (N.C.); (R.C.)
| | - Scott Edwards
- Department of Physiology, Louisiana State University, Health Sciences Center, New Orleans, LA 70112, USA;
| | - Eric P. Zorrilla
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (V.V.); (B.C.); (E.P.Z.)
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy; (F.B.); (N.C.); (R.C.)
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (V.V.); (B.C.); (E.P.Z.)
| |
Collapse
|
16
|
Stopponi S, Fotio Y, Cifani C, Li H, Haass-Koffler CL, Cannella N, Demopulos G, Gaitanaris G, Ciccocioppo R. Andrographis paniculata and Its Main Bioactive Ingredient Andrographolide Decrease Alcohol Drinking and Seeking in Rats Through Activation of Nuclear PPARγ Pathway. Alcohol Alcohol 2021; 56:240-249. [PMID: 33401299 DOI: 10.1093/alcalc/agaa136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND AIMS Andrographis paniculata is an annual herbaceous plant which belongs to the Acanthaceae family. Extracts from this plant have shown hepatoprotective, anti-inflammatory and antidiabetic properties, at least in part, through activation of the nuclear receptor Peroxisome Proliferator-Activated Receptor-gamma (PPAR γ). Recent evidence has demonstrated that activation of PPARγ reduces alcohol drinking and seeking in Marchigian Sardinian (msP) alcohol-preferring rats. METHODS The present study evaluated whether A. paniculata reduces alcohol drinking and relapse in msP rats by activating PPARγ. RESULTS Oral administration of an A. paniculata dried extract (0, 15, 150 mg/kg) lowered voluntary alcohol consumption in a dose-dependent manner and achieved ~65% reduction at the dose of 450 mg/kg. Water and food consumption were not affected by the treatment. Administration of Andrographolide (5 and 10 mg/kg), the main active component of A. paniculata, also reduced alcohol drinking. This effect was suppressed by the selective PPARγ antagonist GW9662. Subsequently, we showed that oral administration of A. paniculata (0, 150, 450 mg/kg) prevented yohimbine- but not cues-induced reinstatement of alcohol seeking. CONCLUSIONS Results point to A. paniculata-mediated PPARγactivation as a possible therapeutic strategy to treat alcohol use disorder.
Collapse
Affiliation(s)
- Serena Stopponi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Yannick Fotio
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy.,Department of Anatomy and Neurobiology, School of Medicine, University of California, 807 Health Science Road, 92617 Irvine, USA
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Hongwu Li
- College of Chemical Engineering, Changchun University of Technology, 2055 Yan An Road, Chao Yang District, 130021 Changchun, China
| | - Carolina L Haass-Koffler
- Center Alcohol and Addiction Studies, Department Psychiatry and Human Behavior Department Behavioral and Social Sciences Brow University 121 S. Main Street, Providence, RI 02931, USA
| | - Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Gregory Demopulos
- Center Alcohol and Addiction Studies, Department Psychiatry and Human Behavior Department Behavioral and Social Sciences Brow University 121 S. Main Street, Providence, RI 02931, USA
| | - George Gaitanaris
- Center Alcohol and Addiction Studies, Department Psychiatry and Human Behavior Department Behavioral and Social Sciences Brow University 121 S. Main Street, Providence, RI 02931, USA.,Omeros Corporation, 201 Elliot Avenue West, Seattle, WA 98119, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| |
Collapse
|
17
|
Walker LC. A balancing act: the role of pro- and anti-stress peptides within the central amygdala in anxiety and alcohol use disorders. J Neurochem 2021; 157:1615-1643. [PMID: 33450069 DOI: 10.1111/jnc.15301] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/18/2020] [Accepted: 01/06/2021] [Indexed: 12/21/2022]
Abstract
The central nucleus of the amygdala (CeA) is widely implicated as a structure that integrates both appetitive and aversive stimuli. While intrinsic CeA microcircuits primarily consist of GABAergic neurons that regulate amygdala output, a notable feature of the CeA is the heterogeneity of neuropeptides and neuropeptide/neuromodulator receptors that it expresses. There is growing interest in the role of the CeA in mediating psychopathologies, including stress and anxiety states and their interactions with alcohol use disorders. Within the CeA, neuropeptides and neuromodulators often exert pro- or anti- stress actions, which can influence anxiety and alcohol associated behaviours. In turn, alcohol use can cause adaptions within the CeA, which may render an individual more vulnerable to stress which is a major trigger of relapse to alcohol seeking. This review examines the neurocircuitry, neurochemical phenotypes and how pro- and anti-stress peptide systems act within the CeA to regulate anxiety and alcohol seeking, focusing on preclinical observations from animal models. Furthermore, literature exploring the targeting of genetically defined populations or neuronal ensembles and the role of the CeA in mediating sex differences in stress x alcohol interactions are explored.
Collapse
Affiliation(s)
- Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Parkville, Vic, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Vic, Australia
| |
Collapse
|
18
|
Abstract
Alcohol dependence is a chronically relapsing disorder characterized by compulsive drug-seeking and drug-taking, loss of control in limiting intake, and the emergence of a withdrawal syndrome in the absence of the drug. Accumulating evidence suggests an important role for synaptic transmission in the central nucleus of the amygdala (CeA) in mediating alcohol-related behaviors and neuroadaptive mechanisms associated with alcohol dependence. Acute alcohol facilitates γ-aminobutyric acid (GABA)ergic transmission in the CeA via both pre- and postsynaptic mechanisms, and chronic alcohol increases baseline GABAergic transmission. Acute alcohol inhibits glutamatergic transmission via effects at N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the CeA, whereas chronic alcohol up-regulates NMDA receptor (NMDAR)-mediated transmission. Pro- (e.g., corticotropin-releasing factor [CRF]) and antistress (e.g., nociceptin/orphanin FQ, oxytocin) neuropeptides affect alcohol- and anxiety-related behaviors, and also alter the alcohol-induced effects on CeA neurotransmission. Alcohol dependence produces plasticity in these neuropeptide systems, reflecting a recruitment of those systems during the transition to alcohol dependence.
Collapse
Affiliation(s)
- Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Dean Kirson
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Sophia Khom
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
19
|
Borruto AM, Stopponi S, Li H, Weiss F, Roberto M, Ciccocioppo R. Genetically selected alcohol-preferring msP rats to study alcohol use disorder: Anything lost in translation? Neuropharmacology 2021; 186:108446. [PMID: 33476639 DOI: 10.1016/j.neuropharm.2020.108446] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/24/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022]
Abstract
For several decades, genetically selected alcohol-preferring rats have been successfully used to mimic and study alcohol use disorders (AUD). These rat lines have been instrumental in advancing our understanding of the neurobiology of alcoholism and enabling pharmacological studies to evaluate drug efficacy on alcohol drinking and relapse. Moreover, the results of these studies have identified genetic variables that are linked to AUD vulnerability. This is an up-to-date review that focuses on genetically selected Marchigian Sardinian alcohol-preferring (msP) rats. To support the translational relevance of the findings that are obtained from msP rats and highlight important similarities to AUD patients, we also discuss the results of recent brain imaging studies. Finally, to demonstrate the importance of studying sex differences in animal models of AUD, we present original data that highlight behavioral differences in the response to alcohol in male and female rats. Female msP rats exhibited higher alcohol consumption compared with males. Furthermore, msP rats of both sexes exhibit higher anxiety- and depressive-like behaviors in the elevated plus maze and forced swim test, respectively, compared with unselected Wistar controls. Notably, voluntary alcohol drinking decreases foot-shock stress and depressive-like behavior in both sexes, whereas anxiety-like behavior in the elevated plus maze is attenuated only in males. These findings suggest that male and female msP rats both drink high amounts of alcohol to self-medicate negative affective symptoms. For females, this behavior may be driven by an attempt to treat stress and depressive-like conditions. For males, generalized anxiety appears to be an important additional factor in the motivation to drink alcohol. This article is part of the special issue on 'Vulnerabilities to Substance Abuse.'
Collapse
Affiliation(s)
- Anna Maria Borruto
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Serena Stopponi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Hongwu Li
- College of Chemical Engineering, Changchun University of Technology, Changchun, China
| | - Friedbert Weiss
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy.
| |
Collapse
|
20
|
N-acylethanolamine acid amidase (NAAA) inhibition decreases the motivation for alcohol in Marchigian Sardinian alcohol-preferring rats. Psychopharmacology (Berl) 2021; 238:249-258. [PMID: 33037452 PMCID: PMC7796956 DOI: 10.1007/s00213-020-05678-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/05/2020] [Indexed: 10/23/2022]
Abstract
RATIONALE N-acylethanolamine acid amidase (NAAA) is an intracellular cysteine hydrolase that terminates the biological actions of oleoylethanolamide (OEA) and palmitoylethanolamide (PEA), two endogenous lipid-derived agonists of the nuclear receptor, and peroxisome proliferator-activated receptor-α. OEA and PEA are important regulators of energy balance, pain, and inflammation, but recent evidence suggests that they might also contribute to the control of reward-related behaviors. OBJECTIVES AND METHODS In the present study, we investigated the effects of systemic and intracerebral NAAA inhibition in the two-bottle choice model of voluntary alcohol drinking and on operant alcohol self-administration. RESULTS Intraperitoneal injections of the systemically active NAAA inhibitor ARN19702 (3 and 10 mg/kg) lowered voluntary alcohol intake in a dose-dependent manner, achieving ≈ 47% reduction at the 10 mg/kg dose (p < 0.001). Water, food, or saccharin consumption was not affected by the inhibitor. Similarly, ARN19702 dose-dependently attenuated alcohol self-administration under both fixed ratio 1 (FR-1) and progressive ratio schedules of reinforcement. Furthermore, microinjection of ARN19702 (1, 3 and 10 μg/μl) or of two chemically different NAAA inhibitors, ARN077 and ARN726 (both at 3 and 10 μg/μl), into the midbrain ventral tegmental area produced dose-dependent decreases in alcohol self-administration under FR-1 schedule. Microinjection of ARN19702 into the nucleus accumbens had no such effect. CONCLUSION Collectively, the results point to NAAA as a possible molecular target for the treatment of alcohol use disorder.
Collapse
|
21
|
Avchalumov Y, Piña-Crespo JC, Woodward JJ, Mandyam CD. Acute Ethanol Exposure Enhances Synaptic Plasticity in the Dorsal Striatum in Adult Male and Female Rats. Brain Plast 2020; 6:113-122. [PMID: 33680850 PMCID: PMC7903017 DOI: 10.3233/bpl-190097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Background: Acute (ex vivo) and chronic (in vivo) alcohol exposure induces neuroplastic changes in the dorsal striatum, a
critical region implicated in instrumental learning. Objective: Sex differences are evident in alcohol reward and reinforcement, with
female rats consuming higher amount of alcohol in operant paradigms compared to male rats. However, sex differences in
the neuroplastic changes produced by acute alcohol in the dorsal striatum have been unexplored. Methods: Using electrophysiological
recordings from dorsal striatal slices obtained from adult male and female rats, we investigated the effects of ex vivo ethanol
exposure on synaptic transmission and synaptic plasticity. Ethanol (44 mM) enhanced basal synaptic transmission in both
sexes. Ethanol also enhanced long-term potentiation in both sexes. Other measures of synaptic plasticity including paired-pulse
ratio were unaltered by ethanol in both sexes. Results: The results suggest that alterations in synaptic plasticity induced by acute
ethanol, at a concentration associated with intoxication, could play an important role in alcohol-induced experience-dependent
modification of corticostriatal circuits underlying the learning of goal-directed instrumental actions and formation of habits
mediating alcohol seeking and taking. Conclusions: Taken together, understanding the mechanism(s) underlying alcohol induced changes
in corticostriatal function may lead to the development of more effective therapeutic agents to reduce habitual drinking and
seeking associated with alcohol use disorders.
Collapse
Affiliation(s)
| | - Juan C Piña-Crespo
- Neuroscience Initiative, Sanford Burnham Prebys Medical Research Institute, La Jolla, CA, USA
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Chitra D Mandyam
- VA San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
22
|
Cucinello-Ragland JA, Edwards S. Neurobiological aspects of pain in the context of alcohol use disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:1-29. [PMID: 33648668 DOI: 10.1016/bs.irn.2020.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Alcohol is an effective and widely utilized analgesic. However, the chronic use of alcohol can actually facilitate nociceptive sensitivity over time, a condition known as hyperalgesia. Excessive and uncontrollable alcohol drinking is also a hallmark feature of alcohol use disorder (AUD). Both AUD and chronic pain are typically accompanied by negative affective states that may underlie reinforcement mechanisms contributing to AUD maintenance or progression. Frequent utilization of alcohol to relieve pain in individuals suffering from AUD or other chronic pain conditions may thus represent a powerful negative reinforcement construct. This chapter will describe ties between alcohol-mediated pain relief and potential exacerbation of AUD. We describe neurobiological systems engaged in alcohol analgesia as well as systems recruited in the development and maintenance of AUD and hyperalgesia. Although few effective therapies exist for either chronic pain or AUD, the common interaction of these conditions will likely lead the way for promising new discoveries of more effective and even simultaneous treatment of AUD and co-morbid hyperalgesia. An abundance of neurobiological findings from multiple laboratories has implicated a potentiation of central amygdala (CeA) signaling in both pain and AUD, and these data also suggest that attenuation of stress-related systems (including corticotropin-releasing factor, vasopressin, and glucocorticoid receptor activity) would be particularly effective and comprehensive therapeutic strategies targeting the critical intersection of somatic and motivational mechanisms driving AUD, including alcohol-induced hyperalgesia.
Collapse
Affiliation(s)
- Jessica A Cucinello-Ragland
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health Sciences Center, New Orleans, LA, United States
| | - Scott Edwards
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health Sciences Center, New Orleans, LA, United States.
| |
Collapse
|
23
|
Avegno EM, Middleton JW, Gilpin NW. Synaptic GABAergic transmission in the central amygdala (CeA) of rats depends on slice preparation and recording conditions. Physiol Rep 2020; 7:e14245. [PMID: 31587506 PMCID: PMC6778595 DOI: 10.14814/phy2.14245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 08/30/2019] [Accepted: 09/05/2019] [Indexed: 11/24/2022] Open
Abstract
The central nucleus of the amygdala (CeA) is a primarily GABAergic brain region implicated in stress and addictive disorders. Using in vitro slice electrophysiology, many studies measure GABAergic neurotransmission to evaluate the impact of experimental manipulations on inhibitory tone in the CeA, as a measure of alterations in CeA activity and function. In a recent study, we reported spontaneous inhibitory postsynaptic current (sIPSC) frequencies higher than those typically reported in CeA neurons in the literature, despite utilizing similar recording protocols and internal recording solutions. The purpose of this study was to systematically evaluate two common methods of slice preparation, an NMDG-based aCSF perfusion method and an ice-cold sucrose solution, as well as the use of an in-line heater to control recording temperature, on measures of intrinsic excitability and spontaneous inhibitory neurotransmission in CeA neurons. We report that both slice preparation and recording conditions significantly impact spontaneous GABAergic transmission in CeA neurons, and that recording temperature, but not slicing solution, alters measures of intrinsic excitability in CeA neurons. Bath application of corticotropin-releasing factor (CRF) increased sIPSC frequency under all conditions, but the magnitude of this effect was significantly different across recording conditions that elicited different baseline GABAergic transmission. Furthermore, CRF effects on synaptic transmission differed according to data reporting methods (i.e., raw vs. normalized data), which is important to consider in relation to baseline synaptic transmission values. These studies highlight the impact of experimental conditions and data reporting methods on neuronal excitability and synaptic transmission in the CeA.
Collapse
Affiliation(s)
- Elizabeth M Avegno
- Department of Physiology, Louisiana State University Health Science Center, New Orleans, Louisiana
| | - Jason W Middleton
- Department of Cell Biology and Anatomy, Louisiana State University Health Science Center, New Orleans, Louisiana.,Department of Neuroscience Center of Excellence, Louisiana State University Health Science Center, New Orleans, Louisiana
| | - Nicholas W Gilpin
- Department of Physiology, Louisiana State University Health Science Center, New Orleans, Louisiana.,Department of Neuroscience Center of Excellence, Louisiana State University Health Science Center, New Orleans, Louisiana
| |
Collapse
|
24
|
Kirson D, Oleata CS, Roberto M. Taurine Suppression of Central Amygdala GABAergic Inhibitory Signaling via Glycine Receptors Is Disrupted in Alcohol Dependence. Alcohol Clin Exp Res 2019; 44:445-454. [PMID: 31782155 DOI: 10.1111/acer.14252] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/18/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Alcohol use disorder (AUD) increases brain stress systems while suppressing reward system functioning. One expression of stress system recruitment is elevated GABAergic activity in the central amygdala (CeA), which is involved in the excessive drinking seen with AUD. The sulfonic amino acid taurine, a glycine receptor partial agonist, modulates GABAergic activity in the rewarding effects of alcohol. Despite taurine abundance in the amygdala, its role in the dysregulation of GABAergic activity associated with AUD has not been studied. Thus, here, we evaluated the effects of taurine on locally stimulated GABAergic neurotransmission in the CeA of naïve- and alcohol-dependent rats. METHODS We recorded intracellularly from CeA neurons of naïve- and alcohol-dependent rats, quantifying locally evoked GABAA receptor-mediated inhibitory postsynaptic potentials (eIPSP). We examined the effects of taurine and alcohol on CeA eIPSP to characterize potential alcohol dependence-induced changes in the effects of taurine. RESULTS We found that taurine decreased amplitudes of eIPSP in CeA neurons of naïve rats, without affecting the acute alcohol-induced facilitation of GABAergic responses. In CeA neurons from dependent rats, taurine no longer had an effect on eIPSP, but now blocked the ethanol (EtOH)-induced increase in eIPSP amplitude normally seen. Additionally, preapplication of the glycine receptor-specific antagonist strychnine blocked the EtOH-induced increase in eIPSP amplitude in neurons from naïve rats. CONCLUSIONS These data suggest taurine may act to oppose the effects of acute alcohol via the glycine receptor in the CeA of naïve rats, and this modulatory system is altered in the CeA of dependent rats.
Collapse
Affiliation(s)
- Dean Kirson
- Department of Molecular Medicine, and Alcohol Research Center, The Scripps Research Institute, La Jolla, California
| | - Christopher S Oleata
- Department of Molecular Medicine, and Alcohol Research Center, The Scripps Research Institute, La Jolla, California
| | - Marisa Roberto
- Department of Molecular Medicine, and Alcohol Research Center, The Scripps Research Institute, La Jolla, California
| |
Collapse
|
25
|
Redlich R, Schneider I, Kerkenberg N, Opel N, Bauhaus J, Enneking V, Repple J, Leehr EJ, Grotegerd D, Kähler C, Förster K, Dohm K, Meinert S, Hahn T, Kugel H, Schwarte K, Schettler C, Domschke K, Arolt V, Heindel W, Baune BT, Zhang W, Hohoff C, Dannlowski U. The role of BDNF methylation and Val 66 Met in amygdala reactivity during emotion processing. Hum Brain Mapp 2019; 41:594-604. [PMID: 31617281 PMCID: PMC7268057 DOI: 10.1002/hbm.24825] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 09/17/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022] Open
Abstract
Epigenetic alterations of the brain-derived neurotrophic factor (BDNF) gene have been associated with psychiatric disorders in humans and with differences in amygdala BDNF mRNA levels in rodents. This human study aimed to investigate the relationship between the functional BDNF-Val66 Met polymorphism, its surrounding DNA methylation in BDNF exon IX, amygdala reactivity to emotional faces, and personality traits. Healthy controls (HC, n = 189) underwent functional MRI during an emotional face-matching task. Harm avoidance, novelty seeking and reward dependence were measured using the Tridimensional Personality Questionnaire (TPQ). Individual BDNF methylation profiles were ascertained and associated with several BDNF single nucleotide polymorphisms surrounding the BDNF-Val66 Met, amygdala reactivity, novelty seeking and harm avoidance. Higher BDNF methylation was associated with higher amygdala reactivity (x = 34, y = 0, z = -26, t(166) = 3.00, TFCE = 42.39, p(FWE) = .045), whereby the BDNF-Val66 Met genotype per se did not show any significant association with brain function. Furthermore, novelty seeking was negatively associated with BDNF methylation (r = -.19, p = .015) and amygdala reactivity (r = -.17, p = .028), while harm avoidance showed a trend for a positive association with BDNF methylation (r = .14, p = .066). The study provides first insights into the relationship among BDNF methylation, BDNF genotype, amygdala reactivity and personality traits in humans, highlighting the multidimensional relations among genetics, epigenetics, and neuronal functions. The present study suggests a possible involvement of epigenetic BDNF modifications in psychiatric disorders and related brain functions, whereby high BDNF methylation might reduce BDNF mRNA expression and upregulate amygdala reactivity.
Collapse
Affiliation(s)
- Ronny Redlich
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Ilona Schneider
- Department of Psychiatry, University of Münster, Münster, Germany.,Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, Münster, Germany
| | | | - Nils Opel
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Jonas Bauhaus
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Verena Enneking
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Jonathan Repple
- Department of Psychiatry, University of Münster, Münster, Germany
| | | | | | - Claas Kähler
- Department of Psychiatry, University of Münster, Münster, Germany
| | | | - Katharina Dohm
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Susanne Meinert
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Tim Hahn
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Harald Kugel
- Department of Clinical Radiology, University of Münster, Münster, Germany
| | - Kathrin Schwarte
- Department of Psychiatry, University of Münster, Münster, Germany
| | | | - Katharina Domschke
- Department of Psychiatry, University of Münster, Münster, Germany.,Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Volker Arolt
- Department of Psychiatry, University of Münster, Münster, Germany.,Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, Münster, Germany
| | - Walter Heindel
- Department of Clinical Radiology, University of Münster, Münster, Germany
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, Münster, Germany.,Department of Psychiatry, Melbourne Medical School and The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Weiqi Zhang
- Department of Psychiatry, University of Münster, Münster, Germany.,Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, Münster, Germany
| | - Christa Hohoff
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Udo Dannlowski
- Department of Psychiatry, University of Münster, Münster, Germany.,Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, Münster, Germany
| |
Collapse
|
26
|
Centanni SW, Bedse G, Patel S, Winder DG. Driving the Downward Spiral: Alcohol-Induced Dysregulation of Extended Amygdala Circuits and Negative Affect. Alcohol Clin Exp Res 2019; 43:2000-2013. [PMID: 31403699 PMCID: PMC6779502 DOI: 10.1111/acer.14178] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/07/2019] [Indexed: 12/17/2022]
Abstract
Alcohol use disorder (AUD) afflicts a large number of individuals, families, and communities globally. Affective disturbances, including stress, depression, and anxiety, are highly comorbid with AUD, contributing in some cases to initial alcohol use and continued use. Negative affect has a particularly strong influence on the withdrawal/abstinence stage of addiction as individuals with AUD frequently report stressful events, depression, and anxiety as key factors for relapse. Treatment options for negative affect associated with AUD are limited and often ineffective, highlighting the pressing need for preclinical studies examining the underlying neural circuitry driving AUD-associated negative affect. The extended amygdala (EA) is a set of brain areas collectively involved in generating and regulating affect, and extensive research has defined a critical role for the EA in all facets of substance use disorder. Here, we review the expansive historical literature examining the effects of ethanol exposure on the EA, with an emphasis on the complex EA neural circuitry driving negative affect in all phases of the alcohol addiction cycle. Specifically, this review focuses on the effects of alcohol exposure on the neural circuitry in 2 key components of the EA, the central nucleus of the amygdala and the bed nucleus of the stria terminalis. Additionally, future directions are proposed to advance our understanding of the relationship between AUD-associated negative affect and neural circuitry in the EA, with the long-term goal of developing better diagnostic tools and new pharmacological targets aimed at treating negative affect in AUD. The concepts detailed here will serve as the foundation for a companion review focusing on the potential for the endogenous cannabinoid system in the EA as a novel target for treating the stress, anxiety, and negative emotional state driving AUD.
Collapse
Affiliation(s)
- Samuel W. Centanni
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
| | - Gaurav Bedse
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Sachin Patel
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Danny G. Winder
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| |
Collapse
|
27
|
Bedse G, Centanni SW, Winder DG, Patel S. Endocannabinoid Signaling in the Central Amygdala and Bed Nucleus of the Stria Terminalis: Implications for the Pathophysiology and Treatment of Alcohol Use Disorder. Alcohol Clin Exp Res 2019; 43:2014-2027. [PMID: 31373708 PMCID: PMC6779484 DOI: 10.1111/acer.14159] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/18/2019] [Indexed: 12/23/2022]
Abstract
High rates of relapse are a chronic and debilitating obstacle to effective treatment of alcohol use disorder (AUD); however, no effective treatments are available to treat symptoms induced by protracted abstinence. In the first part of this 2-part review series, we examine the literature supporting the effects of alcohol exposure within the extended amygdala (EA) neural circuitry. In Part 2, we focus on a potential way to combat negative affect associated with AUD, by exploring the therapeutic potential of the endogenous cannabinoid (eCB) system. The eCB system is a potent modulator of neural activity in the brain, and its ability to mitigate stress and negative affect has long been an area of interest for developing novel therapeutics. This review details the recent advances in our understanding of eCB signaling in 2 key regions of the EA, the central nucleus of the amygdala and the bed nucleus of the stria terminalis (BNST), and their role in regulating negative affect. Despite an established role for EA eCB signaling in reducing negative affect, few studies have examined the potential for eCB-based therapies to treat AUD-associated negative affect. In this review, we present an overview of studies focusing on eCB signaling in EA and cannabinoid modulation on EA synaptic activity. We further discuss studies suggesting dysregulation of eCB signaling in models of AUD and propose that pharmacological augmentation of eCB could be a novel approach to treat aspects of AUD. Lastly, future directions are proposed to advance our understanding of the relationship between AUD-associated negative affect and the EA eCB system that could yield new pharmacotherapies targeting negative affective symptoms associated with AUD.
Collapse
Affiliation(s)
- Gaurav Bedse
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Samuel W. Centanni
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, the, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
| | - Danny G. Winder
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, the, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Sachin Patel
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, the, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| |
Collapse
|
28
|
Abstract
Ethanol produces intoxication through actions on numerous molecular and cellular targets. Adaptations involving these and other targets contribute to chronic drug actions that underlie continued and problematic drinking. Among the mechanisms involved in these ethanol actions are alterations in presynaptic mechanisms of synaptic transmission, including presynaptic protein function and excitation-secretion coupling. At synapses in the central nervous system (CNS), excitation-secretion coupling involves ion channel activation followed by vesicle fusion and neurotransmitter release. These mechanisms are altered by presynaptic neurotransmitter receptors and prominently by G protein-coupled receptors (GPCRs). Studies over the last 20-25 years have revealed that acute ethanol exposure alters neurotransmitter secretion, with especially robust effects on synapses that use the neurotransmitter gamma-aminobutyric acid (GABA). Intracellular signaling pathways involving second messengers such as cyclic AMP and calcium are implicated in these acute ethanol actions. Ethanol-induced release of neuropeptides and small molecule neurotransmitters that act on presynaptic GPCRs also contribute to presynaptic potentiation at synapses in the amygdala and hippocampus and inhibition of GABA release in the striatum. Prolonged exposure to ethanol alters neurotransmitter release at many CNS GABAergic and glutamatergic synapses, and changes in GPCR function are implicated in many of these neuroadaptations. These presynaptic neuroadaptations appear to involve compensation for acute drug effects at some synapses, but "allostatic" effects that result in long-term resetting of synaptic efficacy occur at others. Current investigations are determining how presynaptic neuroadaptations contribute to behavioral changes at different stages of alcohol drinking, with increasing focus on circuit adaptations underlying these behaviors. This chapter will discuss the acute and chronic presynaptic effects of ethanol in the CNS, as well as some of the consequences of these effects in amygdala and corticostriatal circuits that are related to excessive seeking/drinking and ethanol abuse.
Collapse
Affiliation(s)
- David M Lovinger
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA.
| |
Collapse
|
29
|
Endocannabinoid System and Alcohol Abuse Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1162:89-127. [PMID: 31332736 DOI: 10.1007/978-3-030-21737-2_6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Δ9-tetrahydrocannabinol (Δ9-THC), the primary active component in Cannabis sativa preparations such as hashish and marijuana, signals by binding to cell surface receptors. Two types of receptors have been cloned and characterized as cannabinoid (CB) receptors. CB1 receptors (CB1R) are ubiquitously present in the central nervous system (CNS) and are present in both inhibitory interneurons and excitatory neurons at the presynaptic terminal. CB2 receptors (CB2R) are demonstrated in microglial cells, astrocytes, and several neuron subpopulations and are present in both pre- and postsynaptic terminals. The majority of studies on these receptors have been conducted in the past two and half decades after the identification of the molecular constituents of the endocannabinoid (eCB) system that started with the characterization of CB1R. Subsequently, the seminal discovery was made, which suggested that alcohol (ethanol) alters the eCB system, thus establishing the contribution of the eCB system in the motivation to consume ethanol. Several preclinical studies have provided evidence that CB1R significantly contributes to the motivational and reinforcing properties of ethanol and that the chronic consumption of ethanol alters eCB transmitters and CB1R expression in the brain nuclei associated with addiction pathways. Additionally, recent seminal studies have further established the role of the eCB system in the development of ethanol-induced developmental disorders, such as fetal alcohol spectrum disorders (FASD). These results are augmented by in vitro and ex vivo studies, showing that acute and chronic treatment with ethanol produces physiologically relevant alterations in the function of the eCB system during development and in the adult stage. This chapter provides a current and comprehensive review of the literature concerning the role of the eCB system in alcohol abuse disorders (AUD).
Collapse
|
30
|
Logrip ML, Milivojevic V, Bertholomey ML, Torregrossa MM. Sexual dimorphism in the neural impact of stress and alcohol. Alcohol 2018; 72:49-59. [PMID: 30227988 PMCID: PMC6148386 DOI: 10.1016/j.alcohol.2018.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/07/2018] [Accepted: 02/19/2018] [Indexed: 12/18/2022]
Abstract
Alcohol use disorder is a widespread mental illness characterized by periods of abstinence followed by recidivism, and stress is the primary trigger of relapse. Despite the higher prevalence of alcohol use disorder in males, the relationship between stress and behavioral features of relapse, such as craving, is stronger in females. Given the greater susceptibility of females to stress-related psychiatric disorders, understanding sexual dimorphism in the relationship between stress and alcohol use is essential to identifying better treatments for both male and female alcoholics. This review addresses sex differences in the impact of stressors on alcohol drinking and seeking in rodents and humans. As these behavioral differences in alcohol use and relapse originate from sexual dimorphism in neuronal function, the impact of stressors and alcohol, and their interaction, on molecular adaptations and neural activity in males and females will also be discussed. Together, the data reviewed herein, arising from a symposium titled "Sex matters in stress-alcohol interactions" presented at the Fourth Volterra Conference on Stress and Alcohol, will highlight the importance of identifying sex differences to improve treatments for comorbid stress and alcohol use disorder in both sexes.
Collapse
Affiliation(s)
- Marian L Logrip
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States.
| | - Verica Milivojevic
- The Yale Stress Center, Yale University School of Medicine, New Haven, CT 06519, United States
| | - Megan L Bertholomey
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Mary M Torregrossa
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA 15219, United States
| |
Collapse
|
31
|
Wellman CL, Bangasser DA, Bollinger JL, Coutellier L, Logrip ML, Moench KM, Urban KR. Sex Differences in Risk and Resilience: Stress Effects on the Neural Substrates of Emotion and Motivation. J Neurosci 2018; 38:9423-9432. [PMID: 30381434 PMCID: PMC6209838 DOI: 10.1523/jneurosci.1673-18.2018] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/20/2018] [Accepted: 09/22/2018] [Indexed: 02/06/2023] Open
Abstract
Risk for stress-sensitive psychopathologies differs in men and women, yet little is known about sex-dependent effects of stress on cellular structure and function in corticolimbic regions implicated in these disorders. Determining how stress influences these regions in males and females will deepen our understanding of the mechanisms underlying sex-biased psychopathology. Here, we discuss sex differences in CRF regulation of arousal and cognition, glucocorticoid modulation of amygdalar physiology and alcohol consumption, the age-dependent impact of social stress on prefrontal pyramidal cell excitability, stress effects on the prefrontal parvalbumin system in relation to emotional behaviors, contributions of stress and gonadal hormones to stress effects on prefrontal glia, and alterations in corticolimbic structure and function after cessation of chronic stress. These studies demonstrate that, while sex differences in stress effects may be nuanced, nonuniform, and nonlinear, investigations of these differences are nonetheless critical for developing effective, sex-specific treatments for psychological disorders.
Collapse
Affiliation(s)
- Cara L Wellman
- Department of Psychological and Brain Sciences, Program in Neuroscience, and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, Indiana 47405,
| | - Debra A Bangasser
- Psychology Department and Neuroscience Program, Temple University, Philadelphia, Pennsylvania 19122
| | - Justin L Bollinger
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| | - Laurence Coutellier
- Departments of Psychology and Neuroscience, Ohio State University, Columbus, Ohio 43210
| | - Marian L Logrip
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana 46202, and
| | - Kelly M Moench
- Department of Psychological and Brain Sciences, Program in Neuroscience, and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, Indiana 47405
| | - Kimberly R Urban
- Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, Pennsylvania 19104
| |
Collapse
|
32
|
Broccoli L, Uhrig S, von Jonquieres G, Schönig K, Bartsch D, Justice NJ, Spanagel R, Sommer W, Klugmann M, Hansson A. Targeted overexpression of CRH receptor subtype 1 in central amygdala neurons: effect on alcohol-seeking behavior. Psychopharmacology (Berl) 2018; 235:1821-1833. [PMID: 29700576 PMCID: PMC7454014 DOI: 10.1007/s00213-018-4908-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 04/12/2018] [Indexed: 12/14/2022]
Abstract
RATIONALE The corticotropin-releasing hormone (CRH) system is a key mediator of stress-induced responses in alcohol-seeking behavior. Recent research has identified the central nucleus of the amygdala (CeA), a brain region involved in the regulation of fear and stress-induced responses that is especially rich in CRH-positive neurons, as a key player in mediating excessive alcohol seeking. However, detailed characterization of the specific influences that local neuronal populations exert in mediating alcohol responses is hampered by current limitations in pharmacological and immunohistochemical tools for targeting CRH receptor subtype 1 (CRHR1). OBJECTIVE In this study, we investigated the effect of cell- and region-specific overexpression of CRHR1 in the CeA using a novel transgenic tool. METHODS Co-expression of CRHR1 in calcium-calmodulin-dependent kinase II (αCaMKII) neurons of the amygdala was demonstrated by double immunohistochemistry using a Crhr1-GFP reporter mouse line. A Cre-inducible Crhr1-expressing adeno-associated virus (AAV) was site-specifically injected into the CeA of αCaMKII-CreERT2 transgenic rats to analyze the role of CRHR1 in αCaMKII neurons on alcohol self-administration and reinstatement behavior. RESULTS Forty-eight percent of CRHR1-containing cells showed co-expression of αCaMKII in the CeA. AAV-mediated gene transfer in αCaMKII neurons induced a 24-fold increase of Crhr1 mRNA in the CeA which had no effect on locomotor activity, alcohol self-administration, or cue-induced reinstatement. However, rats overexpressing Crhr1 in the CeA increased responding in the stress-induced reinstatement task with yohimbine serving as a pharmacological stressor. CONCLUSION We demonstrate that CRHR1 overexpression in CeA-αCaMKII neurons is sufficient to mediate increased vulnerability to stress-triggered relapse into alcohol seeking.
Collapse
Affiliation(s)
- L. Broccoli
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - S. Uhrig
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - G. von Jonquieres
- Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, UNSW Australia Sydney, NSW, Australia
| | - K. Schönig
- Dept. of Molecular Biology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Square J5, 68159 Mannheim, Germany
| | - D. Bartsch
- Dept. of Molecular Biology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Square J5, 68159 Mannheim, Germany
| | - N. J. Justice
- Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, Texas 77030, USA
| | - R. Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - W.H. Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany,Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, J5, 68159 Mannheim, Germany
| | - M. Klugmann
- Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, UNSW Australia Sydney, NSW, Australia
| | - A.C. Hansson
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany,To whom correspondence should be addressed: Anita C. Hansson, PhD, Institute of Psychopharmacology, Central Institute for Mental Health, University of Heidelberg, Medical Faculty Mannheim, Square J5, D-68159 Mannheim, Germany, Phone: +49 621 1703 6293, Fax: +49 621 1703 6255,
| |
Collapse
|
33
|
Abrahao KP, Salinas AG, Lovinger DM. Alcohol and the Brain: Neuronal Molecular Targets, Synapses, and Circuits. Neuron 2017; 96:1223-1238. [PMID: 29268093 PMCID: PMC6566861 DOI: 10.1016/j.neuron.2017.10.032] [Citation(s) in RCA: 282] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/30/2017] [Accepted: 10/27/2017] [Indexed: 12/13/2022]
Abstract
Ethanol is one of the most commonly abused drugs. Although environmental and genetic factors contribute to the etiology of alcohol use disorders, it is ethanol's actions in the brain that explain (1) acute ethanol-related behavioral changes, such as stimulant followed by depressant effects, and (2) chronic changes in behavior, including escalated use, tolerance, compulsive seeking, and dependence. Our knowledge of ethanol use and abuse thus relies on understanding its effects on the brain. Scientists have employed both bottom-up and top-down approaches, building from molecular targets to behavioral analyses and vice versa, respectively. This review highlights current progress in the field, focusing on recent and emerging molecular, cellular, and circuit effects of the drug that impact ethanol-related behaviors. The focus of the field is now on pinpointing which molecular effects in specific neurons within a brain region contribute to behavioral changes across the course of acute and chronic ethanol exposure.
Collapse
Affiliation(s)
- Karina P Abrahao
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Armando G Salinas
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
34
|
Castelli V, Brancato A, Cavallaro A, Lavanco G, Cannizzaro C. Homer2 and Alcohol: A Mutual Interaction. Front Psychiatry 2017; 8:268. [PMID: 29249995 PMCID: PMC5714871 DOI: 10.3389/fpsyt.2017.00268] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/17/2017] [Indexed: 01/05/2023] Open
Abstract
The past two decades of data derived from addicted individuals and preclinical animal models of addiction implicate a role for the excitatory glutamatergic transmission within the mesolimbic structures in alcoholism. The cellular localization of the glutamatergic receptor subtypes, as well as their signaling efficiency and function, are highly dependent upon discrete functional constituents of the postsynaptic density, including the Homer family of scaffolding proteins. The consequences of repeated alcohol administration on the expression of the Homer family proteins demonstrate a crucial and active role, particularly for the expression of Homer2 isoform, in regulating alcohol-induced behavioral and cellular neuroplasticity. The interaction between Homer2 and alcohol can be defined as a mutual relation: alcohol consumption enhances the expression of Homer2 protein isoform within the nucleus accumbens and the extended amygdala, cerebral areas where, in turn, Homer2 is able to mediate the development of the "pro-alcoholic" behavioral phenotype, as a consequence of the morpho-functional synaptic adaptations. Such findings are relevant for the detection of the strategic molecular components that prompt alcohol-induced functional and behavioral disarrangement as targets for future innovative treatment options.
Collapse
Affiliation(s)
- Valentina Castelli
- Department of Sciences for Health Promotion and Mother and Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Anna Brancato
- Department of Sciences for Health Promotion and Mother and Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Angela Cavallaro
- Department of Sciences for Health Promotion and Mother and Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Gianluca Lavanco
- Department of Sciences for Health Promotion and Mother and Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Carla Cannizzaro
- Department of Sciences for Health Promotion and Mother and Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| |
Collapse
|