1
|
Lonnberg A, Logrip ML, Kuznetsov A. Mechanisms of alcohol influence on fear conditioning: A computational model. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025. [PMID: 40390190 DOI: 10.1111/acer.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/14/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND A connection between stress-related illnesses and alcohol use disorders is extensively documented. Fear conditioning is a standard procedure used to study stress learning and links it to the activation of amygdala circuitry. However, the connection between the changes in amygdala circuitry and function induced by alcohol and fear conditioning is not well established. METHODS We introduce a computational model to test the mechanistic relationship between amygdala functional and circuit adaptations during fear conditioning and the impact of acute vs. repeated alcohol exposure. Using firing rate formalism, the model generates electrophysiological and behavioral responses in fear conditioning protocols via plasticity of amygdala inputs. The influence of alcohol is modeled by accounting for known modulation of connections within amygdala circuits, which consequently affect plasticity. Thus, the model connects the electrophysiological and behavioral experiments. We hypothesize that alterations within amygdala circuitry produced by alcohol cause abnormal plasticity of amygdala inputs such that fear extinction is slower to achieve and less robust. RESULTS In accordance with prior experimental results, both acute and prior repeated alcohol decrease the speed and robustness of fear extinction in our simulations. The model predicts that, first, the delay in fear extinction caused by alcohol is mostly induced by greater activation of the basolateral amygdala (BLA) after fear acquisition due to alcohol-induced modulation of synaptic weights. Second, both acute and prior repeated alcohol shift the amygdala network away from the robust extinction regime by inhibiting activity in the central amygdala (CeA). Third, our model predicts that fear memories formed during acute or after chronic alcohol are more connected to the context. CONCLUSIONS The model suggests how circuit changes induced by alcohol may affect fear behaviors and provides a framework for investigating the involvement of multiple neuromodulators in this neuroadaptive process.
Collapse
Affiliation(s)
- Adam Lonnberg
- Cleveland Clinic, Neurology Residency, Cleveland, Ohio, USA
| | - Marian L Logrip
- Department of Psychology, Indiana University Indianapolis, Indianapolis, Indiana, USA
| | - Alexey Kuznetsov
- Department of Mathematical Sciences, Indiana University Indianapolis, Indianapolis, Indiana, USA
| |
Collapse
|
2
|
Sommer WH, Canals S. Alcohol-Induced Changes in Brain Microstructure: Uncovering Novel Pathophysiological Mechanisms of AUD Using Translational DTI in Humans and Rodents. Curr Top Behav Neurosci 2025. [PMID: 40360929 DOI: 10.1007/7854_2025_585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Alcohol use disorder (AUD) induces significant structural alterations in both gray and white matter, contributing to cognitive and functional impairments. This chapter presents a translational neuroimaging approach using diffusion-weighted MRI in humans and rodents to uncover novel pathophysiological mechanisms underlying AUD. Our studies demonstrate that increased mean diffusivity (MD) in gray matter reflects microglial reactivity and reduced extracellular space tortuosity, leading to enhanced volume neurotransmission. In white matter, fractional anisotropy (FA) reductions indicate progressive deterioration of key tracts, particularly the fimbria/fornix, linked to impaired cognitive flexibility. Importantly, longitudinal analyses reveal that white matter degeneration continues during early abstinence, suggesting that neuroinflammation and demyelination persist beyond alcohol cessation. Finally, we discuss how neuromodulatory interventions, such as transcranial magnetic stimulation (TMS), may promote recovery by enhancing myelin plasticity. These findings provide crucial insights into AUD's neurobiological underpinnings and highlight potential therapeutic targets for improving treatment outcomes.
Collapse
Affiliation(s)
- Wolfgang H Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, Mannheim, Germany.
| | - Santiago Canals
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad Miguel Hernandez (UMH), Sant Joan d'Alacant, Spain.
| |
Collapse
|
3
|
Schank JR, Besheer J, Lovelock DF. Shaken and stirred: Stress and alcohol are a potent mix driving behavior and neuroinflammation. J Pharmacol Exp Ther 2025; 392:103576. [PMID: 40286521 DOI: 10.1016/j.jpet.2025.103576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Affiliation(s)
- Jesse R Schank
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia.
| | - Joyce Besheer
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Dennis F Lovelock
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
4
|
Asrar B, Hassam M, Rafi S, Ullah I, Homberg JR, Haleem DJ. Nigella sativa oil modulates neurobehavioral and neurochemical alterations in alcohol-exposed rats: An in vivo and in silico study. Behav Brain Res 2025; 484:115494. [PMID: 40015344 DOI: 10.1016/j.bbr.2025.115494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 02/08/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Chronic alcohol (ethanol) drinking changes central serotonin and dopamine levels, and thereby the functioning of brain circuits that support cognition and anxiety. Previously, it has been proven that Nigella sativa oil (NSO) improves cognition and reduces anxiety by regulating the neurotransmission but the underlying mechanisms are unknown. METHODS To address the knowledge gap, an in vivo experiment was done to investigate effects of NSO on behavior and neurotransmission in ethanol drinking Wistar male rats. Specifically, control, NSO treated, ethanol and ethanol + NSO treated groups were tested for changes in anxiety-like behavior, locomotor activity and learning and memory using the elevated plus-maze test (EPM) and light and dark (L&D) box test; open field test (OFT) and Morris water maze (MWM) test, respectively. Brain neurotransmitter concentrations were determined using HPLC-EC. To validate the in vivo findings, we assessed in silico the docking between NSO compounds and proteins using auto dock vina. KEY FINDINGS Ethanol and NSO reduced weight in the ethanol and ethanol + NSO groups. Food intake, fluid consumption, calorie intake, and growth were similarly affected by ethanol and NSO. In the in behavioral tests, ethanol drinking rats spent less time in the open arms of the EPM and had fewer entries compared to controls, while ethanol + NSO group also showed reduced entries. Similar patterns were observed in the OFT. No differences were found in the L&D box test. In the memory tests, ethanol + NSO treatment increased latency in short-term memory, while ethanol consumption increased latency in retention. Neurochemical analysis revealed that ethanol + NSO treatment increased serotonin levels in the PFC and hippocampus while reducing dopamine levels in the PFC compared to all groups, and in the hippocampus compared to control and NSO groups. The in silico experiment revealed that NSO has nine main active compounds. By molecular docking, we found that all nine compounds showed good binding affinity with our target proteins but the best docking values were obtained with thymoquinone and dithymoquinone. The binding affinity estimations identified the superior binding affinity and efficiency of dithymoquinone over all nine NSO compounds for serotonin, dopamine receptors and MAO-enzymes. CONCLUSIONS AND SIGNIFICANCE NSO partially modulated ethanol induced neurobehavioral and neurochemical alterations, improving serotonin levels but not fully reversing behavioral deficits. Further studies are needed to explore its protective potential.
Collapse
Affiliation(s)
- Beenish Asrar
- Department of Psychology, Boğaziçi University, Istanbul 34342, Turkey; Sulaiman Bin Abdullah Aba Al-Khail - Centre for Interdisciplinary Research in Basic Science (SA-CIRBS), International Islamic University, Sector H-10, Islamabad, Pakistan; Dr Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Science, (ICCBS), University of Karachi, Karachi 75270, Pakistan; Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Kapittelweg 29, Nijmegen 6525 EN, the Netherlands.
| | - Muhammad Hassam
- Dr Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Science, (ICCBS), University of Karachi, Karachi 75270, Pakistan
| | - Sidra Rafi
- Dr Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Science, (ICCBS), University of Karachi, Karachi 75270, Pakistan; Department of Biological and Biomedical Sciences, Sohail University, Karachi
| | - Ikram Ullah
- Sulaiman Bin Abdullah Aba Al-Khail - Centre for Interdisciplinary Research in Basic Science (SA-CIRBS), International Islamic University, Sector H-10, Islamabad, Pakistan
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Kapittelweg 29, Nijmegen 6525 EN, the Netherlands; Department of Biological and Biomedical Sciences, Sohail University, Karachi
| | - Darakhshan Jabeen Haleem
- Dr Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Science, (ICCBS), University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
5
|
Rawls E, Marquardt CA, Fix ST, Bernat E, Sponheim SR. Posttraumatic reexperiencing and alcohol use: Mediofrontal theta as a neural mechanism for negative reinforcement. JOURNAL OF PSYCHOPATHOLOGY AND CLINICAL SCIENCE 2025; 134:308-318. [PMID: 39899115 PMCID: PMC11949716 DOI: 10.1037/abn0000925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Over half of U.S. military veterans with posttraumatic stress disorder (PTSD) use alcohol heavily, potentially to cope with their symptoms. This study investigated the neural underpinnings of PTSD symptoms and heavy drinking in veterans. We focused on brain responses to salient outcomes within predictive coding theory. This framework suggests the brain generates prediction errors (PEs) when outcomes deviate from expectations. Alcohol use might provide negative reinforcement by reducing the salience of negatively valenced PEs and dampening experiences like loss. We analyzed electroencephalography responses to unpredictable gain/loss feedback in n = 82 veterans of Operations Enduring and Iraqi Freedom. We used time-frequency principal components analysis of event-related potentials to isolate neural responses indicative of PEs, identifying mediofrontal theta linked to losses (feedback-related negativity) and central delta associated with gains (reward positivity). Intrusive reexperiencing symptoms of PTSD were associated with intensified mediofrontal theta signaling during losses, suggesting heightened negative PE sensitivity. Conversely, increased hazardous alcohol use was associated with reduced theta responses, implying a dampening of these negative PEs. The separate delta-reward positivity component showed associations with alcohol use but not PTSD symptoms. The findings suggest a common neural component of PTSD and hazardous alcohol use involving altered PE processing. We suggest that reexperiencing enhances the intensity of salient negative PEs, while chronic alcohol use may reduce their intensity, thereby providing negative reinforcement by muting posttraumatic distress and associated brain responses. Modifying the mediofrontal theta response could address the intertwined nature of PTSD symptoms and alcohol use, providing new avenues for treatment. (PsycInfo Database Record (c) 2025 APA, all rights reserved).
Collapse
Affiliation(s)
- Eric Rawls
- Department of Psychiatry and Behavioral Sciences, University of Minnesota
| | - Craig A. Marquardt
- Department of Psychiatry and Behavioral Sciences, University of Minnesota
- Minneapolis Veterans Affairs Health Care System
| | - Spencer T. Fix
- Department of Psychology, University of Maryland, College Park
| | - Edward Bernat
- Department of Psychology, University of Maryland, College Park
| | - Scott R. Sponheim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota
- Minneapolis Veterans Affairs Health Care System
| |
Collapse
|
6
|
Mayberry HK, Rinker JA, Chandler LJ. Effect of chronic alcohol exposure and single-prolonged stress on conditioned fear behavior. Behav Brain Res 2025; 477:115294. [PMID: 39419182 DOI: 10.1016/j.bbr.2024.115294] [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: 06/19/2024] [Revised: 08/17/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
The present study investigated the impact of chronic intermittent ethanol (CIE) exposure and single-prolonged stress (SPS) on the acquisition of fear memories in both male and female Wistar rats. Adult rats were first subjected to CIE by vapor inhalation followed by SPS. Following a subsequent 8-day incubation period, the rats underwent a Pavlovian fear conditioning procedure (tone-shock pairings) followed by cued-tone extinction training, and then testing of extinction recall memory and fear renewal memory. In control animals that had not been exposed to either CIE or SPS, female rats exhibited significantly lower levels of freezing compared to male rats during tone-shock pairings. This lower level of freezing in female rats during conditioning was associated with an increased speed of movement compared to males. Also compared to males, female rats exhibited lower levels of fear extinction, recall, and renewal. Exposure to CIE, SPS, or CIE+SPS had no effect on freezing during the cued-conditioning, extinction, or extinction recall phases of the testing procedure in either sex. In fear renewal, CIE exposure decreased freezing in male but not female rats, while SPS increased freezing in female but not male rats. CIE exposure significantly reduced freezing during the fear renewal phase. Taken together, these results provide further evidence that male and female rats adopt different avoidance strategies for threat responding. These results also revealed that prior exposure to CIE, SPS, or CIE+SPS had minimal effects on threat responding using conditioned freezing as an indicator of fear responsivity.
Collapse
Affiliation(s)
- Heyam K Mayberry
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Jennifer A Rinker
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - L Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
7
|
Przybysz KR, Shillinglaw JE, Wheeler SR, Glover EJ. Chronic ethanol exposure produces long-lasting, subregion-specific physiological adaptations in RMTg-projecting mPFC neurons. Neuropharmacology 2024; 259:110098. [PMID: 39117106 PMCID: PMC11714651 DOI: 10.1016/j.neuropharm.2024.110098] [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: 05/06/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024]
Abstract
Chronic ethanol exposure produces neuroadaptations in the medial prefrontal cortex (mPFC) that are thought to facilitate maladaptive behaviors that interfere with recovery from alcohol use disorder. Despite evidence that different cortico-subcortical projections play distinct roles in behavior, few studies have examined the physiological effects of chronic ethanol at the circuit level. The rostromedial tegmental nucleus (RMTg) is functionally altered by chronic ethanol exposure. Our recent work identified dense input from the mPFC to the RMTg, yet the effects of chronic ethanol exposure on this circuitry is unknown. In the current study, we examined physiological changes after chronic ethanol exposure in prelimbic (PL) and infralimbic (IL) mPFC neurons projecting to the RMTg. Adult male Long-Evans rats were injected with fluorescent retrobeads into the RMTg and rendered dependent using a 14-day chronic intermittent ethanol (CIE) vapor exposure paradigm. Whole-cell patch-clamp electrophysiological recordings were performed in fluorescently-labeled (RMTg-projecting) and -unlabeled (projection-undefined) layer 5 pyramidal neurons 7-10 days following ethanol exposure. CIE exposure significantly increased intrinsic excitability as well as spontaneous excitatory and inhibitory postsynaptic currents (sE/IPSCs) in RMTg-projecting IL neurons. In contrast, no lasting changes in excitability were observed in RMTg-projecting PL neurons, although a CIE-induced reduction in excitability was observed in projection-undefined PL neurons. CIE exposure also increased the frequency of sEPSCs in RMTg-projecting PL neurons. These data uncover novel subregion- and circuit-specific neuroadaptations in the mPFC following chronic ethanol exposure and reveal that the IL mPFC-RMTg projection is uniquely vulnerable to long-lasting effects of chronic ethanol exposure. This article is part of the Special Issue on "PFC circuit function in psychiatric disease and relevant models".
Collapse
Affiliation(s)
- Kathryn R Przybysz
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois Chicago, Chicago, IL, USA
| | - Joel E Shillinglaw
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois Chicago, Chicago, IL, USA
| | - Shannon R Wheeler
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois Chicago, Chicago, IL, USA
| | - Elizabeth J Glover
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
8
|
Domi A, Cadeddu D, Lucente E, Gobbo F, Edvardsson C, Petrella M, Jerlhag E, Ericson M, Söderpalm B, Adermark L. Pre- and postsynaptic signatures in the prelimbic cortex associated with "alcohol use disorder" in the rat. Neuropsychopharmacology 2024; 49:1851-1860. [PMID: 38755284 PMCID: PMC11473806 DOI: 10.1038/s41386-024-01887-2] [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: 01/29/2024] [Revised: 04/20/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
The transition to alcohol use disorder (AUD) involves persistent neuroadaptations in executive control functions primarily regulated by the medial prefrontal cortex. However, the neurophysiological correlates to behavioral manifestations of AUD are not fully defined. The association between cortical neuroadaptations and behavioral manifestations of addiction was studied using a multi-symptomatic operant model based on the DSM-5 diagnostic criteria for AUD. This model aimed to characterize an AUD-vulnerable and AUD-resistant subpopulation of outbred male Wistar rats and was combined with electrophysiological measurements in the prelimbic cortex (PL). Mirroring clinical observations, rats exhibited individual variability in their vulnerability to develop AUD-like behavior, including motivation to seek for alcohol (crit 1), increased effort to obtain the substance (crit 2), and continued drinking despite negative consequences (crit 3). Only a small subset of rats met all the aforementioned AUD criteria (3 crit, AUD-vulnerable), while a larger fraction was considered AUD-resilient (0 crit). The development of AUD-like behavior was characterized by disruptions in glutamatergic synaptic activity, involving decreased frequency of spontaneous excitatory postsynaptic currents (sEPSCs) and heightened intrinsic excitability in layers 2/3 PL pyramidal neurons. These alterations were concomitant with a significant impairment in the ability of mGlu2/3 receptors to negatively regulate glutamate release in the PL but not in downstream regions like the basolateral amygdala or nucleus accumbens core. In conclusion alterations in PL synaptic activity were strongly associated with individual addiction scores, indicating their role as potential markers of the behavioral manifestations linked to AUD psychopathology.
Collapse
Affiliation(s)
- Ana Domi
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 90, Sweden.
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 45, Sweden.
| | - Davide Cadeddu
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 90, Sweden
| | - Erika Lucente
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 90, Sweden
| | - Francesco Gobbo
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Christian Edvardsson
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 90, Sweden
| | - Michele Petrella
- Linköping University, Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience. Linköping University, Faculty of Medicine and Health Sciences, Linköping, Sweden
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 90, Sweden
| | - Mia Ericson
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 90, Sweden
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 45, Sweden
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 45, Sweden
- The Clinic for Addiction and Dependency, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Louise Adermark
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 90, Sweden
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 45, Sweden
| |
Collapse
|
9
|
Cardona-Jordan KM, Lay-Rivera XX, Cartagena-López E, Bracho-Rincón DL, González-Bermejo R, Alvarado-Monefeldt GL, Del Toro JPG, Esquilín-Rodríguez CJ, Lloret-Torres M, Velázquez-Marrero C. Sex Differences in Contextual Extinction Learning After Single Binge-Like EtOH Exposure in Adolescent C57BL/6J Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620195. [PMID: 39484582 PMCID: PMC11527338 DOI: 10.1101/2024.10.25.620195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The relationship between chronic heavy drinking and post-traumatic stress disorder (PTSD) is well-documented; however, the impact of more common drinking patterns, such as a single episode leading to a blood alcohol concentration (BAC) of 0.09 g/dL (moderate intoxication), remains underexplored. Given the frequent co-occurrence of PTSD and alcohol misuse, it is essential to understand the biological and behavioral factors driving this comorbidity. We hypothesize that alcohol's immediate sedative effects are coupled with the development of persistent molecular alcohol tolerance, which may disrupt fear extinction learning. To investigate this, we employed a S ingle E pisode E thanol (SEE) in-vivo exposure to mimic binge-like alcohol consumption over a 6-hour period, following contextual conditioning trials. Extinction trials were conducted 24 hours later to assess the effects on extinction learning. Our findings reveal a significant deficit in fear extinction learning in alcohol-treated adolescent male mice compared to saline-treated controls, with no such effects observed in female adolescent mice. These results suggest that even non-chronic alcohol exposure may contribute to the development of trauma- and stress-related disorders, such as PTSD, in males. Additionally, histological analysis revealed significant alterations in FKBP5, β-catenin, and GSK-3β levels in the hippocampus, striatum, and basolateral amygdala of alcohol-treated mice following extinction. The insights gained from this study could reshape our understanding of the risk factors for PTSD and open new avenues for prevention and treatment, targeting the molecular mechanisms that mediate alcohol tolerance.
Collapse
Affiliation(s)
- Kiara M Cardona-Jordan
- University of Puerto Rico, Medical Sciences Campus, Dr. Jose Celso Barbosa, San Juan, PR, 00936
| | - Xiany X Lay-Rivera
- University of Puerto Rico, Medical Sciences Campus, Dr. Jose Celso Barbosa, San Juan, PR, 00936
| | - Eliezer Cartagena-López
- Institute of Neurobiology, UPR-Medical Sciences Campus, 201 Blvd del Valle, San Juan, PR, 00901
| | - Dina L Bracho-Rincón
- Neuroimaging and Electrophysiology Facility - Institute of Neurobiology, 201 Blvd del Valle, San Juan, PR, 00901
| | - Ruth González-Bermejo
- Institute of Neurobiology, UPR-Medical Sciences Campus, 201 Blvd del Valle, San Juan, PR, 00901
| | | | | | | | - Mario Lloret-Torres
- University of Puerto Rico, Medical Sciences Campus, Dr. Jose Celso Barbosa, San Juan, PR, 00936
| | | |
Collapse
|
10
|
Hartsock MJ, Levy CT, Navarro MJ, Saddoris MP, Spencer RL. Circadian Rhythms in Conditioned Threat Extinction Reflect Time-of-Day Differences in Ventromedial Prefrontal Cortex Neural Processing. J Neurosci 2024; 44:e0878242024. [PMID: 39251355 PMCID: PMC11426375 DOI: 10.1523/jneurosci.0878-24.2024] [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: 05/06/2024] [Revised: 07/25/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024] Open
Abstract
Circadian rhythms in conditioned threat extinction emerge from a tissue-level circadian timekeeper, or local clock, in the ventromedial prefrontal cortex (vmPFC). Yet it remains unclear how this local clock contributes to extinction-dependent adaptations. Here we used single-unit and local field potential analyses to interrogate neural activity in the male rat vmPFC during repeated extinction sessions at different times of day. In association with superior recall of a remote extinction memory during the circadian active phase, vmPFC putative principal neurons exhibited phasic firing that was amplified for cue presentations and diminished at transitions in freezing behavior. Coupling of vmPFC gamma amplitude to the phase of low-frequency oscillations was greater during freezing than mobility, and this difference was augmented during the active phase, highlighting a time-of-day dependence in the organization of freezing- versus mobility-associated cell assemblies. Additionally, a greater proportion of vmPFC neurons were phase-locked to low-frequency oscillations during the active phase, consistent with heightened neural excitability at this time of day. Our results suggest that daily fluctuations in vmPFC excitability precipitate enhanced neural recruitment into extinction-based cell assemblies during the active phase, providing a potential mechanism by which the vmPFC local clock modulates circuit and behavioral plasticity during conditioned threat extinction.
Collapse
Affiliation(s)
- Matthew J Hartsock
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado 80301
| | - Catherine T Levy
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado 80301
| | - Maria J Navarro
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado 80301
| | - Michael P Saddoris
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado 80301
| | - Robert L Spencer
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado 80301
| |
Collapse
|
11
|
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
|
12
|
Canto-de-Souza L, Baptista-de-Souza D, Nunes-de-Souza RL, Planeta C. Distinct roles of the left and right prelimbic cortices in the modulation of ethanol consumption in male mice under acute and chronic social defeat stress. Psychopharmacology (Berl) 2024; 241:1161-1176. [PMID: 38347153 DOI: 10.1007/s00213-024-06550-8] [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: 11/01/2023] [Accepted: 01/31/2024] [Indexed: 05/21/2024]
Abstract
RATIONALE Chronic stress exposure disrupts the medial prefrontal cortex's (mPFC) ability to regulate impulses, leading to the loss of control over alcohol drinking in rodents, emphasizing the critical role of this forebrain area in regulating alcohol consumption. Moreover, chronic stress exposure causes lateralization of mPFC functions with volumetric and functional changes, resulting in hyperactivity in the right hemisphere and functional decrease in the left. OBJECTIVES This study investigated the inhibitory role of the left prelimbic cortex (LPrL) on ethanol consumption induced by chronic social defeat stress (SDS) in male mice and to examine if inactivation of the LPrL causes disinhibition of the right mPFC, leading to an increase in ethanol consumption. We also investigated the role of lateralization and neurochemical alterations in the mPFC related to ethanol consumption induced by chronic SDS. To this end, we examined the activation patterns of ΔFosB, VGLUT2, and GAD67 in the left and right mPFC. RESULTS Temporarily blocking the LPrL or right PrL (RPrL) cortices during acute SDS did not affect male mice's voluntary ethanol consumption in male mice. When each cortex was blocked in mice previously exposed to chronic SDS, ethanol consumption also remained unaffected. However, male mice with LPrL lesions during chronic SDS showed an increase in voluntary ethanol consumption, which was associated with enhanced ΔFosB/VGLUT2-positive neurons within the RPrL cortex. CONCLUSIONS The results suggest that the LPrL may play a role in inhibiting ethanol consumption induced by chronic SDS, while the RPrL may be involved in the disinhibition of ethanol consumption.
Collapse
Affiliation(s)
- Lucas Canto-de-Souza
- Lab. Pharmacology, School of Pharmaceutical Sciences, São Paulo State University - UNESP, Araraquara, SP, 14800-903, Brazil
| | - Daniela Baptista-de-Souza
- Lab. Pharmacology, School of Pharmaceutical Sciences, São Paulo State University - UNESP, Araraquara, SP, 14800-903, Brazil
| | - Ricardo Luiz Nunes-de-Souza
- Lab. Pharmacology, School of Pharmaceutical Sciences, São Paulo State University - UNESP, Araraquara, SP, 14800-903, Brazil
- Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, SP, 13565-905, Brazil
| | - Cleopatra Planeta
- Lab. Pharmacology, School of Pharmaceutical Sciences, São Paulo State University - UNESP, Araraquara, SP, 14800-903, Brazil.
- Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, SP, 13565-905, Brazil.
| |
Collapse
|
13
|
Kale MB, Chandurkar PA, Taksande BG, Aglawe MM, Rahangdale SR, Upaganlawar AB, Kopalli SR, Umekar MJ, Wankhede NL. Agmatine alleviates ethanol withdrawal-associated cognitive impairment and neurochemical imbalance in rats. Neurosci Lett 2024; 832:137804. [PMID: 38692559 DOI: 10.1016/j.neulet.2024.137804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/03/2024]
Abstract
The present study aimed to investigate the role of agmatine in the neurobiology underlying memory impairment during ethanol withdrawal in rats. Sprague-Dawley rats were subjected to a 21-day chronic ethanol exposure regimen (2.4 % w/v ethanol for 3 days, 4.8 % w/v for the next 4 days, and 7.2 % w/v for the following 14 days), followed by a withdrawal period. Memory impairment was assessed using the passive avoidance test (PAT) at 24, 48, and 72 h post-withdrawal. The ethanol-withdrawn rats displayed a significant decrease in step-through latency in the PAT, indicative of memory impairment at 72 h post-withdrawal. However, administration of agmatine (40 µg/rat) and its modulators (L-arginine, arcaine, and amino-guanidine) significantly increases the latency time in the ethanol-withdrawn rats, demonstrating the attenuation of memory impairment. Further, pretreatment with imidazoline receptor agonists enhances agmatine's effects, while antagonists block them, implicating imidazoline receptors in agmatine's actions. Neurochemical analysis in ethanol-withdrawn rats reveals dysregulated glutamate and GABA levels, which was attenuated by agmatine and its modulators. By examining the effects of agmatine administration and modulators of endogenous agmatine, the study aimed to shed light on the potential therapeutic implications of agmatinergic signaling in alcohol addiction and related cognitive deficits. Thus, the present findings suggest that agmatine administration and modulation of endogenous agmatine levels hold potential as therapeutic strategies for managing alcohol addiction and associated cognitive deficits. Understanding the neurobiology underlying these effects paves the way for the development of novel interventions targeting agmatinergic signaling in addiction treatment.
Collapse
Affiliation(s)
- Mayur B Kale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India; SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra 423101, India.
| | - Pranali A Chandurkar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Brijesh G Taksande
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Manish M Aglawe
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Sandip R Rahangdale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra 423101, India.
| | - Spandana R Kopalli
- Department of Integrated Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea.
| | - Milind J Umekar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Nitu L Wankhede
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India; SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra 423101, India.
| |
Collapse
|
14
|
Przybysz KR, Shillinglaw JE, Wheeler SR, Glover EJ. Chronic ethanol exposure produces long-lasting, subregion-specific physiological adaptations in RMTg-projecting mPFC neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592759. [PMID: 38766178 PMCID: PMC11100703 DOI: 10.1101/2024.05.06.592759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Chronic ethanol exposure produces neuroadaptations in the medial prefrontal cortex (mPFC) which facilitate the maladaptive behaviors interfering with recovery from alcohol use disorder. Despite evidence that different cortico-subcortical projections play distinct roles in behavior, few studies have examined the physiological effects of chronic ethanol at the circuit level. The rostromedial tegmental nucleus (RMTg) is a GABAergic midbrain region involved in aversive signaling and is functionally altered by chronic ethanol exposure. Our recent work identified a dense input from the mPFC to the RMTg, yet the effects of chronic ethanol exposure on this circuitry is unknown. In the current study, we examined physiological changes after chronic ethanol exposure in prelimbic (PL) and infralimbic (IL) mPFC neurons projecting to the RMTg. Adult male Long-Evans rats were injected with fluorescent retrobeads into the RMTg and rendered dependent using a 14-day chronic intermittent ethanol (CIE) vapor exposure paradigm. Whole-cell patch-clamp electrophysiological recordings were performed in fluorescently-labeled (RMTg-projecting) and -unlabeled (projection-undefined) layer 5 pyramidal neurons 7-10 days following ethanol exposure. CIE significantly increased intrinsic excitability as well as excitatory and inhibitory synaptic drive in RMTg-projecting IL neurons. In contrast, no lasting changes in excitability were observed in RMTg-projecting PL neurons, although a CIE-induced reduction in excitability was observed in projection-undefined PL neurons. CIE also increased excitatory synaptic drive in RMTg-projecting PL neurons. These data uncover novel subregion- and circuit-specific neuroadaptations in the mPFC following chronic ethanol exposure and reveal that the IL mPFC-RMTg projection is uniquely vulnerable to long-lasting effects of chronic ethanol.
Collapse
|
15
|
Steiner NL, Purohit DC, Tiefenthaler CM, Mandyam CD. Abstinence and Fear Experienced during This Period Produce Distinct Cortical and Hippocampal Adaptations in Alcohol-Dependent Rats. Brain Sci 2024; 14:431. [PMID: 38790410 PMCID: PMC11118749 DOI: 10.3390/brainsci14050431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/14/2024] [Accepted: 04/20/2024] [Indexed: 05/26/2024] Open
Abstract
Previous studies demonstrate that ethanol dependence induced by repeating cycles of chronic intermittent ethanol vapor exposure (CIE) followed by protracted abstinence produces significant gray matter damage via myelin dysfunction in the rodent medial prefrontal cortex (mPFC) and alterations in neuronal excitability in the mPFC and the dentate gyrus (DG) of the hippocampus. Specifically, abstinence-induced neuroadaptations have been associated with persistent elevated relapse to drinking. The current study evaluated the effects of forced abstinence for 1 day (d), 7 d, 21 d, and 42 d following seven weeks of CIE on synaptic plasticity proteins in the mPFC and DG. Immunoblotting revealed reduced expression of CaMKII in the mPFC and enhanced expression of GABAA and CaMKII in the DG at the 21 d time point, and the expression of the ratio of GluN2A/2B subunits did not change at any of the time points studied. Furthermore, cognitive performance via Pavlovian trace fear conditioning (TFC) was evaluated in 3 d abstinent rats, as this time point is associated with negative affect. In addition, the expression of the ratio of GluN2A/2B subunits and a 3D structural analysis of neurons in the mPFC and DG were evaluated in 3 d abstinent rats. Behavioral analysis revealed faster acquisition of fear responses and reduced retrieval of fear memories in CIE rats compared to controls. TFC produced hyperplasticity of pyramidal neurons in the mPFC under control conditions and this effect was not evident or blunted in abstinent rats. Neurons in the DG were unaltered. TFC enhanced the GluN2A/2B ratio in the mPFC and reduced the ratio in the DG and was not altered by abstinence. These findings indicate that forced abstinence from CIE produces distinct and divergent alterations in plasticity proteins in the mPFC and DG. Fear learning-induced changes in structural plasticity and proteins contributing to it were more profound in the mPFC during forced abstinence.
Collapse
Affiliation(s)
- Noah L. Steiner
- VA San Diego Healthcare System, San Diego, CA 92161, USA; (N.L.S.)
| | | | - Casey M. Tiefenthaler
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA 92093, USA;
| | - Chitra D. Mandyam
- VA San Diego Healthcare System, San Diego, CA 92161, USA; (N.L.S.)
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA 92093, USA;
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92093, USA
| |
Collapse
|
16
|
Shen XY, Zhang J, Huang HZ, Li SD, Zhou L, Wu SP, Tang C, Huang X, Liu ZQ, Guo ZY, Li X, Man HY, Lu YM, Zhu LQ, Liu D. The interaction of Synapsin 2a and Synaptogyrin-3 regulates fear extinction in mice. J Clin Invest 2024; 134:e172802. [PMID: 38175724 PMCID: PMC10866652 DOI: 10.1172/jci172802] [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: 06/05/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
The mechanisms behind a lack of efficient fear extinction in some individuals are unclear. Here, by employing a principal components analysis-based approach, we differentiated the mice into extinction-resistant and susceptible groups. We determined that elevated synapsin 2a (Syn2a) in the infralimbic cortex (IL) to basolateral amygdala (BLA) circuit disrupted presynaptic orchestration, leading to an excitatory/inhibitory imbalance in the BLA region and causing extinction resistance. Overexpression or silencing of Syn2a levels in IL neurons replicated or alleviated behavioral, electrophysiological, and biochemical phenotypes in resistant mice. We further identified that the proline-rich domain H in the C-terminus of Syn2a was indispensable for the interaction with synaptogyrin-3 (Syngr3) and demonstrated that disrupting this interaction restored extinction impairments. Molecular docking revealed that ritonavir, an FDA-approved HIV drug, could disrupt Syn2a-Syngr3 binding and rescue fear extinction behavior in Syn2a-elevated mice. In summary, the aberrant elevation of Syn2a expression and its interaction with Syngr3 at the presynaptic site were crucial in fear extinction resistance, suggesting a potential therapeutic avenue for related disorders.
Collapse
Affiliation(s)
- Xi-Ya Shen
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Juan Zhang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - He-Zhou Huang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shao-Dan Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Zhou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shi-Ping Wu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Cheng Tang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xian Huang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhi-Qiang Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zi-Yuan Guo
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Xiang Li
- Department of Neurosurgery and
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Medical Research Institute, Wuhan University, Wuhan, Hubei, China
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, Massachusetts, USA
| | - You-Ming Lu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dan Liu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
17
|
Lonnberg A, Logrip ML, Kuznetsov A. Mechanisms of alcohol influence on fear conditioning: a computational model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.30.573310. [PMID: 38260700 PMCID: PMC10802259 DOI: 10.1101/2023.12.30.573310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
A connection between stress-related illnesses and alcohol use disorders is extensively documented. Fear conditioning is a standard procedure used to study stress learning and links it to the activation of amygdala circuitry. However, the connection between the changes in amygdala circuit and function induced by alcohol and fear conditioning is not well established. We introduce a computational model to test the mechanistic relationship between amygdala functional and circuit adaptations during fear conditioning and the impact of acute vs. repeated alcohol exposure. In accordance with experiments, both acute and prior repeated alcohol decreases speed and robustness of fear extinction in our simulations. The model predicts that, first, the delay in fear extinction in alcohol is mostly induced by greater activation of the basolateral amygdala (BLA) after fear acquisition due to alcohol-induced modulation of synaptic weights. Second, both acute and prior repeated alcohol shifts the amygdala network away from the robust extinction regime by inhibiting the activity in the central amygdala (CeA). Third, our model predicts that fear memories formed in acute or after chronic alcohol are more connected to the context. Thus, the model suggests how circuit changes induced by alcohol may affect fear behaviors and provides a framework for investigating the involvement of multiple neuromodulators in this neuroadaptive process.
Collapse
Affiliation(s)
- Adam Lonnberg
- University of Evansville, Department of Mathematics, Indianapolis, Indiana, USA
| | - Marian L. Logrip
- Indiana University-Purdue University, Department of Psychology, Indianapolis, Indiana, USA
| | - Alexey Kuznetsov
- Indiana University-Purdue University, Department of Mathematical Sciences, Indianapolis, Indiana, USA
| |
Collapse
|
18
|
Rawls E, Marquardt CA, Fix ST, Bernat E, Sponheim SR. Posttraumatic reexperiencing and alcohol use: mediofrontal theta as a neural mechanism for negative reinforcement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.12.547253. [PMID: 37502872 PMCID: PMC10370024 DOI: 10.1101/2023.07.12.547253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Objective Over half of US military veterans with posttraumatic stress disorder (PTSD) use alcohol heavily, potentially to cope with their symptoms. This study investigated the neural underpinnings of PTSD symptoms and heavy drinking in veterans. We focused on brain responses to salient outcomes within predictive coding theory. This framework suggests the brain generates prediction errors (PEs) when outcomes deviate from expectations. Alcohol use might provide negative reinforcement by reducing the salience of negatively-valenced PEs and dampening experiences like loss. Methods We analyzed electroencephalography (EEG) responses to unpredictable gain/loss feedback in veterans of Operations Enduring and Iraqi Freedom. We used time-frequency principal components analysis of event-related potentials to isolate neural responses indicative of PEs, identifying mediofrontal theta linked to losses (feedback-related negativity, FRN) and central delta associated with gains (reward positivity, RewP). Results Intrusive reexperiencing symptoms of PTSD were associated with intensified mediofrontal theta signaling during losses, suggesting heightened negative PE sensitivity. Conversely, increased hazardous alcohol use was associated with reduced theta responses, implying a dampening of these negative PEs. The separate delta-RewP component showed associations with alcohol use but not PTSD symptoms. Conclusions Findings suggest a common neural component of PTSD and hazardous alcohol use involving altered PE processing. We suggest that reexperiencing enhances the intensity of salient negative PEs, while chronic alcohol use may reduce their intensity, thereby providing negative reinforcement by muting emotional disruption from reexperienced trauma. Modifying the mediofrontal theta response could address the intertwined nature of PTSD symptoms and alcohol use, providing new avenues for treatment.
Collapse
Affiliation(s)
- Eric Rawls
- Department of Psychiatry and Behavioral Sciences, University of Minnesota
| | - Craig A Marquardt
- Minneapolis Veterans Affairs Health Care System
- Department of Psychiatry and Behavioral Sciences, University of Minnesota
| | - Spencer T Fix
- Department of Psychology, University of Maryland College Park
| | - Edward Bernat
- Department of Psychology, University of Maryland College Park
| | - Scott R Sponheim
- Minneapolis Veterans Affairs Health Care System
- Department of Psychiatry and Behavioral Sciences, University of Minnesota
| |
Collapse
|
19
|
Siddiqi MT, Podder D, Pahng AR, Athanason AC, Nadav T, Cates-Gatto C, Kreifeldt M, Contet C, Roberts AJ, Edwards S, Roberto M, Varodayan FP. Prefrontal cortex glutamatergic adaptations in a mouse model of alcohol use disorder. ADDICTION NEUROSCIENCE 2023; 9:100137. [PMID: 38152067 PMCID: PMC10752437 DOI: 10.1016/j.addicn.2023.100137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Alcohol use disorder (AUD) produces cognitive deficits, indicating a shift in prefrontal cortex (PFC) function. PFC glutamate neurotransmission is mostly mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type ionotropic receptors (AMPARs); however preclinical studies have mostly focused on other receptor subtypes. Here we examined the impact of early withdrawal from chronic ethanol on AMPAR function in the mouse medial PFC (mPFC). Dependent male C57BL/6J mice were generated using the chronic intermittent ethanol vapor-two bottle choice (CIE-2BC) paradigm. Non-dependent mice had access to water and ethanol bottles but did not receive ethanol vapor. Naïve mice had no ethanol exposure. We used patch-clamp electrophysiology to measure glutamate neurotransmission in layer 2/3 prelimbic mPFC pyramidal neurons. Since AMPAR function can be impacted by subunit composition or plasticity-related proteins, we probed their mPFC expression levels. Dependent mice had higher spontaneous excitatory postsynaptic current (sEPSC) amplitude and kinetics compared to the Naïve/Non-dependent mice. These effects were seen during intoxication and after 3-8 days withdrawal, and were action potential-independent, suggesting direct enhancement of AMPAR function. Surprisingly, 3 days withdrawal decreased expression of genes encoding AMPAR subunits (Gria1/2) and synaptic plasticity proteins (Dlg4 and Grip1) in Dependent mice. Further analysis within the Dependent group revealed a negative correlation between Gria1 mRNA levels and ethanol intake. Collectively, these data establish a role for mPFC AMPAR adaptations in the glutamatergic dysfunction associated with ethanol dependence. Future studies on the underlying AMPAR plasticity mechanisms that promote alcohol reinforcement, seeking, drinking and relapse behavior may help identify new targets for AUD treatment.
Collapse
Affiliation(s)
- Mahum T. Siddiqi
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Dhruba Podder
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Amanda R. Pahng
- Department of Physiology, Louisiana State University Health Sciences Center, 533 Bolivar Street, New Orleans, LA, 70112, USA
- Southeast Louisiana Veterans Health Care System, 2400 Canal Street, 11F, New Orleans, LA, 70119, USA
| | - Alexandria C. Athanason
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Tali Nadav
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Chelsea Cates-Gatto
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Max Kreifeldt
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Candice Contet
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, 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
| | - Scott Edwards
- Department of Physiology, Louisiana State University Health Sciences Center, 533 Bolivar Street, New Orleans, LA, 70112, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Florence P. Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|
20
|
Reinwald JR, Schmitz CN, Skorodumov I, Kuchar M, Weber-Fahr W, Spanagel R, Meinhardt MW. Psilocybin-induced default mode network hypoconnectivity is blunted in alcohol-dependent rats. Transl Psychiatry 2023; 13:392. [PMID: 38097569 PMCID: PMC10721862 DOI: 10.1038/s41398-023-02690-1] [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: 08/04/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/17/2023] Open
Abstract
Alcohol Use Disorder (AUD) adversely affects the lives of millions of people, but still lacks effective treatment options. Recent advancements in psychedelic research suggest psilocybin to be potentially efficacious for AUD. However, major knowledge gaps remain regarding (1) psilocybin's general mode of action and (2) AUD-specific alterations of responsivity to psilocybin treatment in the brain that are crucial for treatment development. Here, we conducted a randomized, placebo-controlled crossover pharmaco-fMRI study on psilocybin effects using a translational approach with healthy rats and a rat model of alcohol relapse. Psilocybin effects were quantified with resting-state functional connectivity using data-driven whole-brain global brain connectivity, network-based statistics, graph theory, hypothesis-driven Default Mode Network (DMN)-specific connectivity, and entropy analyses. Results demonstrate that psilocybin induced an acute wide-spread decrease in different functional connectivity domains together with a distinct increase of connectivity between serotonergic core regions and cortical areas. We could further provide translational evidence for psilocybin-induced DMN hypoconnectivity reported in humans. Psilocybin showed an AUD-specific blunting of DMN hypoconnectivity, which strongly correlated to the alcohol relapse intensity and was mainly driven by medial prefrontal regions. In conclusion, our results provide translational validity for acute psilocybin-induced neural effects in the rodent brain. Furthermore, alcohol relapse severity was negatively correlated with neural responsivity to psilocybin treatment. Our data suggest that a clinical standard dose of psilocybin may not be sufficient to treat severe AUD cases; a finding that should be considered for future clinical trials.
Collapse
Affiliation(s)
- Jonathan R Reinwald
- Research Group Translational Imaging, Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Research Group Systems Neuroscience and Mental Health, Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Christian N Schmitz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Ivan Skorodumov
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Martin Kuchar
- Forensic Laboratory of Biologically Active Substances, Department of Chemistry of Natural Compounds, University of Chemistry and Technology Prague, Prague, Czech Republic
- Psychedelics Research Centre, National Institute of Mental Health, Klecany, Czech Republic
| | - Wolfgang Weber-Fahr
- Research Group Translational Imaging, Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
| | - Marcus W Meinhardt
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
21
|
Nonoguchi HA, Jin M, Narreddy R, Kouo TWS, Nayak M, Trenet W, Mandyam CD. Progenitor Cells Play a Role in Reinstatement of Ethanol Seeking in Adult Male and Female Ethanol Dependent Rats. Int J Mol Sci 2023; 24:12233. [PMID: 37569609 PMCID: PMC10419311 DOI: 10.3390/ijms241512233] [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: 03/15/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Female and male glial fibrillary acidic protein-thymidine kinase (GFAP-TK) transgenic rats were made ethanol dependent via a six-week chronic intermittent ethanol vapor (CIE) and ethanol drinking (ED) procedure. During the last week of CIE, a subset of male and female TK rats was fed valcyte to ablate dividing progenitor cells and continued the diet until the end of this study. Following week six, all CIE rats experienced two weeks of forced abstinence from CIE-ED, after which they experienced relapse to drinking, extinction, and reinstatement of ethanol seeking sessions. CIE increased ED in female and male rats, with females having higher ethanol consumption during CIE and relapse sessions compared with males. In both sexes, valcyte reduced the levels of Ki-67-labeled progenitor cells in the subgranular zone of the dentate gyrus and did not alter the levels in the medial prefrontal cortex (mPFC). Valcyte increased ED during relapse, increased lever responses during extinction and, interestingly, enhanced latency to extinguish ethanol-seeking behaviors in males. Valcyte reduced the reinstatement of ethanol-seeking behaviors triggered by ethanol cues in females and males. Reduced seeking by valcyte was associated with the normalization of cytokines and chemokines in plasma isolated from trunk blood, indicating a role for progenitor cells in peripheral inflammatory responses. Reduced seeking by valcyte was associated with increases in tight junction protein claudin-5 and oligodendrogenesis in the dentate gyrus and reduction in microglial activity in the dentate gyrus and mPFC in females and males, demonstrating a role for progenitor cells in the dentate gyrus in dependence-induced endothelial and microglial dysfunction. These data suggest that progenitor cells born during withdrawal and abstinence from CIE in the dentate gyrus are aberrant and could play a role in strengthening ethanol memories triggered by ethanol cues via central and peripheral immune responses.
Collapse
Affiliation(s)
| | - Michael Jin
- VA San Diego Healthcare System, San Diego, CA 92161, USA
| | | | | | | | - Wulfran Trenet
- VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Chitra D. Mandyam
- VA San Diego Healthcare System, San Diego, CA 92161, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| |
Collapse
|
22
|
Cucinello-Ragland JA, Alrashed NH, Lee S, Davis EC, Edwards KN, Edwards S. Sex-specific biobehavioral regulation of persistent inflammatory pain by alcohol. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1283-1296. [PMID: 37208939 PMCID: PMC10422981 DOI: 10.1111/acer.15104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/19/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Although a large percentage of chronic pain patients consume alcohol to manage their pain, there is a significant gap in knowledge regarding the mechanisms underlying the antinociceptive effects of alcohol. METHODS To determine the longitudinal analgesic effects of alcohol, we utilized the complete Freund's adjuvant (CFA) model of inflammatory pain in adult female and male Wistar rats. Both somatic and negative motivational aspects of pain were measured using the electronic von Frey (mechanical nociception) system, thermal probe test (thermal nociception), and mechanical conflict avoidance task (pain avoidance-like behavior). Tests were conducted at baseline and 1 and 3 weeks following intraplantar CFA or saline administration. At both time points post-CFA, animals were treated with each of three doses of alcohol (intraperitoneal; 0, 0.5, and 1.0 g/kg) over separate days in a Latin square design. RESULTS Alcohol produced dose-dependent mechanical analgesia and antihyperalgesia in females but only antihyperalgesia in males. Although alcohol continued to attenuate CFA-induced decreases in both thermal and mechanical nociceptive thresholds between 1 and 3 weeks post-CFA, it appeared less effective at increasing thresholds 3 weeks after CFA induction. CONCLUSIONS These data suggest that individuals may develop tolerance to alcohol's ability to alleviate both somatic and negative motivational symptoms of chronic pain over time. We also discovered sex-specific neuroadaptations in protein kinase A-dependent phosphorylation of GluR1 subunits and extracellular signal-regulated kinase (ERK 1/2) phosphorylation in nociceptive brain centers of animals receiving an alcohol challenge 1 week post-CFA. Together, these findings illustrate a sex-specific regulation of behavioral and neurobiological indices of persistent pain by alcohol.
Collapse
Affiliation(s)
- Jessica A. Cucinello-Ragland
- Department of Physiology, LSU Health-New Orleans
- Alcohol & Drug Abuse Center of Excellence, LSU Health-New Orleans
| | | | - Sumin Lee
- Department of Physiology, LSU Health-New Orleans
| | | | | | - Scott Edwards
- Department of Physiology, LSU Health-New Orleans
- Alcohol & Drug Abuse Center of Excellence, LSU Health-New Orleans
- Neuroscience Center of Excellence, LSU Health-New Orleans
- Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health-New Orleans
| |
Collapse
|
23
|
Pérez-Cervera L, De Santis S, Marcos E, Ghorbanzad-Ghaziany Z, Trouvé-Carpena A, Selim MK, Pérez-Ramírez Ú, Pfarr S, Bach P, Halli P, Kiefer F, Moratal D, Kirsch P, Sommer WH, Canals S. Alcohol-induced damage to the fimbria/fornix reduces hippocampal-prefrontal cortex connection during early abstinence. Acta Neuropathol Commun 2023; 11:101. [PMID: 37344865 PMCID: PMC10286362 DOI: 10.1186/s40478-023-01597-8] [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: 05/09/2023] [Accepted: 05/30/2023] [Indexed: 06/23/2023] Open
Abstract
INTRODUCTION Alcohol dependence is characterized by a gradual reduction in cognitive control and inflexibility to contingency changes. The neuroadaptations underlying this aberrant behavior are poorly understood. Using an animal model of alcohol use disorders (AUD) and complementing diffusion-weighted (dw)-MRI with quantitative immunohistochemistry and electrophysiological recordings, we provide causal evidence that chronic intermittent alcohol exposure affects the microstructural integrity of the fimbria/fornix, decreasing myelin basic protein content, and reducing the effective communication from the hippocampus (HC) to the prefrontal cortex (PFC). Using a simple quantitative neural network model, we show how disturbed HC-PFC communication may impede the extinction of maladaptive memories, decreasing flexibility. Finally, combining dw-MRI and psychometric data in AUD patients, we discovered an association between the magnitude of microstructural alteration in the fimbria/fornix and the reduction in cognitive flexibility. Overall, these findings highlight the vulnerability of the fimbria/fornix microstructure in AUD and its potential contribution to alcohol pathophysiology. Fimbria vulnerability to alcohol underlies hippocampal-prefrontal cortex dysfunction and correlates with cognitive impairment.
Collapse
Affiliation(s)
- Laura Pérez-Cervera
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant, Alicante, Spain
| | - Silvia De Santis
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant, Alicante, Spain
| | - Encarni Marcos
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant, Alicante, Spain
| | - Zahra Ghorbanzad-Ghaziany
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant, Alicante, Spain
- Radiation Science and Biomedical Imaging, University of Sherbrooke, Sherbrooke, Québec, Canada
| | - Alejandro Trouvé-Carpena
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant, Alicante, Spain
| | - Mohamed Kotb Selim
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant, Alicante, Spain
| | - Úrsula Pérez-Ramírez
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Simone Pfarr
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Patrick Bach
- Department of Addiction Medicine, Department of Clinical Psychology, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Patrick Halli
- Department of Psychology, University of Heidelberg, Heidelberg, Germany
| | - Falk Kiefer
- Department of Addiction Medicine, Department of Clinical Psychology, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - David Moratal
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Peter Kirsch
- Department of Psychology, University of Heidelberg, Heidelberg, Germany
| | - Wolfgang H Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical faculty Mannheim, University of Heidelberg, Mannheim, Germany.
- Department of Addiction Medicine, Department of Clinical Psychology, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany.
| | - Santiago Canals
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant, Alicante, Spain.
| |
Collapse
|
24
|
McNally GP, Jean-Richard-Dit-Bressel P, Millan EZ, Lawrence AJ. Pathways to the persistence of drug use despite its adverse consequences. Mol Psychiatry 2023; 28:2228-2237. [PMID: 36997610 PMCID: PMC10611585 DOI: 10.1038/s41380-023-02040-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 04/01/2023]
Abstract
The persistence of drug taking despite its adverse consequences plays a central role in the presentation, diagnosis, and impacts of addiction. Eventual recognition and appraisal of these adverse consequences is central to decisions to reduce or cease use. However, the most appropriate ways of conceptualizing persistence in the face of adverse consequences remain unclear. Here we review evidence that there are at least three pathways to persistent use despite the negative consequences of that use. A cognitive pathway for recognition of adverse consequences, a motivational pathway for valuation of these consequences, and a behavioral pathway for responding to these adverse consequences. These pathways are dynamic, not linear, with multiple possible trajectories between them, and each is sufficient to produce persistence. We describe these pathways, their characteristics, brain cellular and circuit substrates, and we highlight their relevance to different pathways to self- and treatment-guided behavior change.
Collapse
Affiliation(s)
- Gavan P McNally
- School of Psychology, UNSW Sydney, Sydney, NSW, 2052, Australia.
| | | | - E Zayra Millan
- School of Psychology, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3010, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3010, Australia
| |
Collapse
|
25
|
Foster TC. Animal models for studies of alcohol effects on the trajectory of age-related cognitive decline. Alcohol 2023; 107:4-11. [PMID: 35504438 DOI: 10.1016/j.alcohol.2022.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023]
Abstract
There is growing interest in understanding how ethanol use interacts with advancing age to influence the brain and cognition. Animal models are employed to investigate the cellular and molecular mechanisms of brain aging and age-related neurodegenerative diseases that underlie cognitive decline. However, all too often research on problems and diseases of the elderly are conducted in healthy young animals, providing little clinical relevance. The validity of animal models is discussed, and confounds due to age-related differences in anxiety, sensory-motor function, and procedural learning are highlighted in order to enhance the successful translation of preclinical results into clinical settings. The mechanism of action of ethanol on brain aging will depend on the dose, acute or chronic treatment, or withdrawal from treatment and the age examined. Due to the fact that humans experience alcohol use throughout life, important questions concern the effects of the dose and duration of ethanol treatment on the trajectory of cognitive function. Central to this research will be questions of the specificity of alcohol effects on cognitive functions and related brain regions that decline with age, as well as the interaction of alcohol with mechanisms or biomarkers of brain aging. Alternatively, moderate alcohol use may provide a source of reserve and resilience against brain aging. Longitudinal studies have the advantage of being sensitive to detecting the effects of treatment on the emergence of cognitive impairment in middle age and can minimize effects of stress/anxiety associated with the novelty of alcohol exposure and behavioral testing, which disproportionately influence aged animals. Finally, the effect of alcohol on senescent neurophysiology and biomarkers of brain aging are discussed. In particular, the interaction of age and effects of alcohol on inflammation, oxidative stress, N-methyl-d-aspartate receptor function, and the balance of excitatory and inhibitory synaptic transmission are highlighted.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
26
|
Hua SS, Ding JJ, Sun TC, Guo C, Zhang Y, Yu ZH, Cao YQ, Zhong LH, Wu Y, Guo LY, Luo JH, Cui YH, Qiu S. NMDAR-dependent synaptic potentiation via APPL1 signaling is required for the accessibility of a prefrontal neuronal assembly in retrieving fear extinction. Biol Psychiatry 2023:S0006-3223(23)00087-2. [PMID: 36842495 DOI: 10.1016/j.biopsych.2023.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/02/2023] [Accepted: 02/14/2023] [Indexed: 02/26/2023]
Abstract
BACKGROUND The ventromedial prefrontal cortex (vmPFC) has been viewed as a locus to store and recall extinction memory. However, the synaptic and cellular mechanisms underlying this process remain elusive. METHODS We combined transgenic mice, electrophysiological recording, activity-dependent cell labeling, and chemogenetic manipulation to analyze the role of adaptor protein APPL1 in the vmPFC for fear extinction retrieval. RESULTS We found that both constitutive and conditional APPL1 knockout decreases NMDA receptor (NMDAR) function in the vmPFC and impairs fear extinction retrieval. Moreover, APPL1 undergoes nuclear translocation during extinction retrieval. Blocking APPL1 nucleocytoplasmic translocation reduces NMDAR currents and disrupts extinction retrieval. We further identified a prefrontal neuronal ensemble that is both necessary and sufficient for the storage of extinction memory. Inducible APPL1 knockout in this ensemble abolishes NMDAR-dependent synaptic potentiation and disrupts extinction retrieval, while simultaneously chemogenetic activation of this ensemble rescues the impaired behaviors. CONCLUSIONS Therefore, our results indicate that a prefrontal neuronal ensemble stores extinction memory, and APPL1 signaling supports these neurons to retrieve extinction memory via controlling NMDAR-dependent potentiation.
Collapse
Affiliation(s)
- Shu-Shan Hua
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jin-Jun Ding
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Tian-Cheng Sun
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Chen Guo
- Department of Neurobiology and Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ying Zhang
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zi-Hui Yu
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yi-Qing Cao
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Lin-Hong Zhong
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yu Wu
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Lu-Ying Guo
- Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jian-Hong Luo
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology,ZhejiangUniversity ,Hangzhou ,310058 ,China
| | - Yi-Hui Cui
- Department of Neurobiology and Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Shuang Qiu
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology,ZhejiangUniversity ,Hangzhou ,310058 ,China.
| |
Collapse
|
27
|
Gimenez-Gomez P, Le T, Martin GE. Modulation of neuronal excitability by binge alcohol drinking. Front Mol Neurosci 2023; 16:1098211. [PMID: 36866357 PMCID: PMC9971943 DOI: 10.3389/fnmol.2023.1098211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/19/2023] [Indexed: 02/16/2023] Open
Abstract
Drug use poses a serious threat to health systems throughout the world. The number of consumers rises every year being alcohol the drug of abuse most consumed causing 3 million deaths (5.3% of all deaths) worldwide and 132.6 million disability-adjusted life years. In this review, we present an up-to-date summary about what is known regarding the global impact of binge alcohol drinking on brains and how it affects the development of cognitive functions, as well as the various preclinical models used to probe its effects on the neurobiology of the brain. This will be followed by a detailed report on the state of our current knowledge of the molecular and cellular mechanisms underlying the effects of binge drinking on neuronal excitability and synaptic plasticity, with an emphasis on brain regions of the meso-cortico limbic neurocircuitry.
Collapse
Affiliation(s)
- Pablo Gimenez-Gomez
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
| | - Timmy Le
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, United States
| | - Gilles E. Martin
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- The Brudnick Neuropsychiatric Research Institute, Worcester, MA, United States
| |
Collapse
|
28
|
Zhan B, Zhu Y, Xia J, Li W, Tang Y, Beesetty A, Ye JH, Fu R. Comorbidity of Post-Traumatic Stress Disorder and Alcohol Use Disorder: Animal Models and Associated Neurocircuitry. Int J Mol Sci 2022; 24:ijms24010388. [PMID: 36613829 PMCID: PMC9820348 DOI: 10.3390/ijms24010388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/12/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) and alcohol use disorder (AUD) are prevalent neuropsychiatric disorders and frequently co-occur concomitantly. Individuals suffering from this dual diagnosis often exhibit increased symptom severity and poorer treatment outcomes than those with only one of these diseases. Lacking standard preclinical models limited the exploration of neurobiological mechanisms underlying PTSD and AUD comorbidity. In this review, we summarize well-accepted preclinical model paradigms and criteria for developing successful models of comorbidity. We also outline how PTSD and AUD affect each other bidirectionally in the nervous nuclei have been heatedly discussed recently. We hope to provide potential recommendations for future research.
Collapse
Affiliation(s)
- Bo Zhan
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
| | - Yingxin Zhu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
| | - Jianxun Xia
- Department of Basic Medical Sciences, Yunkang School of Medicine and Health, Nanfang College, Guangzhou 510970, China
| | - Wenfu Li
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
| | - Ying Tang
- Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Anju Beesetty
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ 07103, USA
| | - Jiang-Hong Ye
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ 07103, USA
- Correspondence: (J.-H.Y.); (R.F.)
| | - Rao Fu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
- Correspondence: (J.-H.Y.); (R.F.)
| |
Collapse
|
29
|
Jiang W, Chen J, Vidjro OE, Zhang Y, Guo G, Li Z, Qi Y, Dai R, Ma T. Construction and evaluation of an alcohol vapor chamber system. J Biomed Res 2022; 37:115-124. [PMID: 36529969 PMCID: PMC10018410 DOI: 10.7555/jbr.36.20220151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
An increasing number of studies demonstrated that alcohol vapor chamber is an effective way to model physical signs of alcohol use disorders. Although researchers are developing different vapor chambers to study chronic alcohol exposure model worldwide, few studies build and modify their own vapor chambers in China. Here, we designed and established an alcohol vapor chamber system for small animals. We described a paradigm showing how to control and monitor alcohol concentration in whole system. The vapor chamber system with several advantages including accommodating up to ten standard mouse cages. Furthermore, the system was tested by evaluating the blood alcohol concentration and neuron injury in mice. Importantly, the alcohol withdrawal after vapor exposure caused motor coordination impairment, anxiolytic- and depression-like behavior. Finally, the N-methyl-D-aspartate receptor (NMDAR)-mediated glutamatergic transmissions in the medial prefrontal cortex was changed after alcohol vapor exposure-induced behaviors. The frequency and amplitude of spontaneous excitatory postsynaptic currents between control and alcohol groups were not different, suggesting that alcohol exposure-induced behaviors are associated with the change in NMDAR response. Taken together, the new alcohol vapor chamber system was constructed, which would help to research the relationship between the stable alcohol exposure and withdrawal behaviors and to study chronic alcohol exposure-induced disorders in China.
Collapse
Affiliation(s)
- Wan Jiang
- Grade 2018, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiajia Chen
- Grade 2018, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Olivia Ewi Vidjro
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Translational Medicine Research Center for Drug Dependence and Withdrawal, Nanjing Medical University, Jiangsu 211166, China
| | - Yingying Zhang
- Grade 2018, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Gengni Guo
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ziyi Li
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Translational Medicine Research Center for Drug Dependence and Withdrawal, Nanjing Medical University, Jiangsu 211166, China
| | - Yize Qi
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Translational Medicine Research Center for Drug Dependence and Withdrawal, Nanjing Medical University, Jiangsu 211166, China
| | - Rouli Dai
- National Institute of Drug Clinical Trial, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210031, China
| | - Tengfei Ma
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Translational Medicine Research Center for Drug Dependence and Withdrawal, Nanjing Medical University, Jiangsu 211166, China
| |
Collapse
|
30
|
Lawson K, Scarlata MJ, Cho WC, Mangan C, Petersen D, Thompson HM, Ehnstrom S, Mousley AL, Bezek JL, Bergstrom HC. Adolescence alcohol exposure impairs fear extinction and alters medial prefrontal cortex plasticity. Neuropharmacology 2022; 211:109048. [PMID: 35364101 PMCID: PMC9067297 DOI: 10.1016/j.neuropharm.2022.109048] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/26/2022] [Accepted: 03/26/2022] [Indexed: 10/18/2022]
Abstract
After experiencing a traumatic event people often turn to alcohol to cope with symptoms. In those with post-traumatic stress disorder (PTSD) and a co-occurring alcohol use disorder (AUD), PTSD symptoms can worsen, suggesting that alcohol changes how traumatic memory is expressed. The objective of this series of experiments is to identify how alcohol drinking (EtOH), following cued fear conditioning and extinction, impacts fear expression in mice. Molecular (activity-regulated cytoskeleton-associated protein, Arc/arg3.1) and structural (dendrite and spine morphometry) markers of neuronal plasticity were measured following remote extinction retrieval. Mouse age (adolescent and adult) and sex were included as interacting variables in a full factorial design. Females drank more EtOH than males and adolescents drank more EtOH than adults. Adolescent females escalated EtOH intake across drinking days. Adolescent drinkers exhibited more conditioned freezing during extinction retrieval, an effect that persisted for at least 20 days. Heightened cued freezing in the adolescent group was associated with greater Arc/arg3.1 expression in layer (L) 2/3 prelimbic (PL) cortex, greater spine density, and reduced basal dendrite complexity. In adults, drinking was associated with reduced L2/3 infralimbic (IL) Arc expression but no behavioral differences. Few sex interactions were uncovered throughout. Overall, these data identify prolonged age-related differences in alcohol-induced fear extinction impairment and medial prefrontal cortex neuroadaptations.
Collapse
Affiliation(s)
- K Lawson
- Department of Psychological Science, Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, USA
| | - M J Scarlata
- Department of Psychological Science, Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, USA
| | - W C Cho
- Department of Psychological Science, Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, USA
| | - C Mangan
- Department of Psychological Science, Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, USA
| | - D Petersen
- Department of Psychological Science, Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, USA
| | - H M Thompson
- Department of Psychological Science, Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, USA
| | - S Ehnstrom
- Department of Psychological Science, Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, USA
| | - A L Mousley
- Department of Psychological Science, Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, USA
| | - J L Bezek
- Department of Psychological Science, Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, USA
| | - H C Bergstrom
- Department of Psychological Science, Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY, USA.
| |
Collapse
|
31
|
Bloch S, Holleran KM, Kash TL, Vazey EM, Rinker JA, Lebonville CL, O'Hara K, Lopez MF, Jones SR, Grant KA, Becker HC, Mulholland PJ. Assessing negative affect in mice during abstinence from alcohol drinking: Limitations and future challenges. Alcohol 2022; 100:41-56. [PMID: 35181404 PMCID: PMC8983487 DOI: 10.1016/j.alcohol.2022.02.001] [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] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 01/09/2023]
Abstract
Alcohol use disorder (AUD) is frequently comorbid with mood disorders, and these co-occurring neuropsychiatric disorders contribute to the development and maintenance of alcohol dependence and relapse. In preclinical models, mice chronically exposed to alcohol display anxiety-like and depressive-like behaviors during acute withdrawal and protracted abstinence. However, in total, results from studies using voluntary alcohol-drinking paradigms show variable behavioral outcomes in assays measuring negative affective behaviors. Thus, the main objective of this review is to summarize the literature on the variability of negative affective behaviors in mice after chronic alcohol exposure. We compare the behavioral phenotypes that emerge during abstinence across different exposure models, including models of alcohol and stress interactions. The complicated outcomes from these studies highlight the difficulties of assessing negative affective behaviors in mouse models designed for the study of AUD. We discuss new behavioral assays, comprehensive platforms, and unbiased machine-learning algorithms as promising approaches to better understand the interaction between alcohol and negative affect in mice. New data-driven approaches in the understanding of mouse behavior hold promise for improving the identification of mechanisms, cell subtypes, and neurocircuits that mediate negative affect. In turn, improving our understanding of the neurobehavioral basis of alcohol-associated negative affect will provide a platform to test hypotheses in mouse models that aim to improve the development of more effective strategies for treating individuals with AUD and co-occurring mood disorders.
Collapse
Affiliation(s)
- Solal Bloch
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Katherine M Holleran
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Elena M Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003, United States
| | - Jennifer A Rinker
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Christina L Lebonville
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Krysten O'Hara
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Marcelo F Lopez
- Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States
| | - Howard C Becker
- Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States.
| |
Collapse
|
32
|
Wei G, Sirohi S, Walker BM. Dysregulated kappa-opioid receptors in the medial prefrontal cortex contribute to working memory deficits in alcohol dependence. Addict Biol 2022; 27:e13138. [PMID: 35138672 PMCID: PMC8829053 DOI: 10.1111/adb.13138] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 01/14/2023]
Abstract
Impaired working memory is one symptom contributing to compromised executive function in alcohol use disorder (AUD). Dysregulation of cortical dynorphin (DYN) and κ-opioid receptors (KORs) has been implicated in alcohol dependence-induced impairment in executive function. The present experiments test the hypothesis that dysregulated medial prefrontal cortex (mPFC) KORs contribute to impaired working memory in alcohol dependence. Alcohol dependence was induced in male Wistar rats via 4 months of intermittent ethanol vapor exposure prior to training/testing in an mPFC-dependent working memory task (delayed nonmatching-to-sample task; DNMST). mPFC KOR function in alcohol-naïve rats was compared with that of alcohol-dependent and nondependent rats using a DYN A-stimulated [35S ]GTPγS coupling assay. A functional role for mPFC KORs in the regulation of working memory was assessed via intra-mPFC infusions of a KOR agonist prior to assessment in the DNMST, and the contribution of mPFC KORs to compromised working memory in dependence was assessed via mPFC infusions of the KOR antagonist norbinaltorphimine (nor-BNI). In alcohol-dependent rats, impaired performance in the DNMST confirmed compromised working memory. Furthermore, DYN A-stimulated mPFC KOR function was pathologically increased in alcohol-dependent rats compared with nondependent and alcohol-naïve rats. Additionally, mPFC KOR involvement in working memory was functionally confirmed by intra-mPFC KOR agonist-induced deficits in DNMST performance. Importantly, alcohol dependence-induced impairment in the DNMST was ameliorated by intra-mPFC KOR antagonism. Regulation of working memory by mPFC KORs and alcohol dependence-induced dysregulation of mPFC KOR function identify a novel therapeutic target to treat AUD-related symptoms of working memory impairment.
Collapse
Affiliation(s)
- Gengze Wei
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Sunil Sirohi
- Laboratory of Endocrine and Neuropsychiatric Disorders, Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA
| | - Brendan M Walker
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL.,Correspondence: Brendan M. Walker, Ph.D., Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychiatry & Behavioral Neurosciences, University of South Florida, 3515 E. Fletcher, Tampa, FL 33613, 813-974-3715 (office),
| |
Collapse
|
33
|
Miczek KA, DiLeo A, Newman EL, Akdilek N, Covington HE. Neurobiological Bases of Alcohol Consumption After Social Stress. Curr Top Behav Neurosci 2022; 54:245-281. [PMID: 34964935 PMCID: PMC9698769 DOI: 10.1007/7854_2021_273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The urge to seek and consume excessive alcohol is intensified by prior experiences with social stress, and this cascade can be modeled under systematically controlled laboratory conditions in rodents and non-human primates. Adaptive coping with intermittent episodes of social defeat stress often transitions to maladaptive responses to traumatic continuous stress, and alcohol consumption may become part of coping responses. At the circuit level, the neural pathways subserving stress coping intersect with those for alcohol consumption. Increasingly discrete regions and connections within the prefrontal cortex, the ventral and dorsal striatum, thalamic and hypothalamic nuclei, tegmental areas as well as brain stem structures begin to be identified as critical for reacting to and coping with social stress while seeking and consuming alcohol. Several candidate molecules that modulate signals within these neural connections have been targeted in order to reduce excessive drinking and relapse. In spite of some early clinical failures, neuropeptides such as CRF, opioids, or oxytocin continue to be examined for their role in attenuating stress-escalated drinking. Recent work has focused on neural sites of action for peptides and steroids, most likely in neuroinflammatory processes as a result of interactive effects of episodic social stress and excessive alcohol seeking and drinking.
Collapse
Affiliation(s)
- Klaus A. Miczek
- Department of Psychology, Tufts University, Medford, MA, USA,Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Alyssa DiLeo
- Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Emily L. Newman
- Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - Naz Akdilek
- Department of Psychology, Tufts University, Medford, MA, USA
| | | |
Collapse
|
34
|
Staples MC, Herman MA, Lockner JW, Avchalumov Y, Kharidia KM, Janda KD, Roberto M, Mandyam CD. Isoxazole-9 reduces enhanced fear responses and retrieval in ethanol-dependent male rats. J Neurosci Res 2021; 99:3047-3065. [PMID: 34496069 PMCID: PMC10112848 DOI: 10.1002/jnr.24932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/30/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022]
Abstract
Plasticity in the dentate gyrus (DG) is strongly influenced by ethanol, and ethanol experience alters long-term memory consolidation dependent on the DG. However, it is unclear if DG plasticity plays a role in dysregulation of long-term memory consolidation during abstinence from chronic ethanol experience. Outbred male Wistar rats experienced 7 weeks of chronic intermittent ethanol vapor exposure (CIE). Seventy-two hours after CIE cessation, CIE and age-matched ethanol-naïve Air controls experienced auditory trace fear conditioning (TFC). Rats were tested for cue-mediated retrieval in the fear context either twenty-four hours (24 hr), ten days (10 days), or twenty-one days (21 days) later. CIE rats showed enhanced freezing behavior during TFC acquisition compared to Air rats. Air rats showed significant fear retrieval, and this behavior did not differ at the three time points. In CIE rats, fear retrieval increased over time during abstinence, indicating an incubation in fear responses. Enhanced retrieval at 21 days was associated with reduced structural and functional plasticity of ventral granule cell neurons (GCNs) and reduced expression of synaptic proteins important for neuronal plasticity. Systemic treatment with the drug Isoxazole-9 (Isx-9; small molecule that stimulates DG plasticity) during the last week and a half of CIE blocked altered acquisition and retrieval of fear memories in CIE rats during abstinence. Concurrently, Isx-9 modulated the structural and functional plasticity of ventral GCNs and the expression of synaptic proteins in the ventral DG. These findings identify that abstinence-induced disruption of fear memory consolidation occurs via altered plasticity within the ventral DG, and that Isx-9 prevented these effects.
Collapse
Affiliation(s)
| | - Melissa A. Herman
- Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jonathan W. Lockner
- Departments of Chemistry and Immunology, Scripps Research, La Jolla, CA, USA
| | | | | | - Kim D. Janda
- Departments of Chemistry and Immunology, Scripps Research, La Jolla, CA, USA
| | - Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, Scripps Research, La Jolla, CA, 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
|
35
|
Smiley CE, McGonigal JT, Nimchuk KE, Gass JT. Optogenetic manipulation of the prelimbic cortex during fear memory reconsolidation alters fear extinction in a preclinical model of comorbid PTSD/AUD. Psychopharmacology (Berl) 2021; 238:3193-3206. [PMID: 34347171 DOI: 10.1007/s00213-021-05935-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/12/2021] [Indexed: 12/27/2022]
Abstract
RATIONALE AND OBJECTIVE Post-traumatic stress disorder (PTSD) and alcohol use disorder (AUD) are disorders of learning and memory that often occur comorbidly. Exposure to trauma-related cues can increase alcohol intake in PTSD patients that are using alcohol to self-medicate. The recurrence of anxiety symptoms with subsequent alcohol use may initiate a destructive cycle where stress and alcohol exposure impair the function of the prefrontal cortex (PFC). While the incidence of these disorders has steadily increased, current therapies and treatments often lack efficacy. Thus, investigation into the underlying neurocircuitry responsible for the establishment and maintenance of these disorders is necessary to develop novel treatment targets. METHODS The present study examined the effects of ethanol exposure on the ability to create new learned associations around previously conditioned fear cues in a rat model. Animals were exposed to fear conditioning followed by chronic intermittent ethanol to translationally model trauma exposure followed by alcohol abuse. Optogenetics was used to inhibit the prelimbic (PrL) or infralimbic (IfL) cortex during fear memory reconsolidation, and fear behaviors were measured during subsequent extinction and spontaneous recovery tests. Results and conclusion Chronic ethanol exposure led to deficits in fear extinction learning and increased freezing during spontaneous recovery, both of which were prevented following inhibition of the PrL, but not the IfL, during memory reconsolidation. These results support the involvement of the PrL in fear learning and memory, and strongly suggest that the PrL could serve as a potential target for the treatment of the learning and memory deficits that occur following exposure to stress and alcohol.
Collapse
Affiliation(s)
- C E Smiley
- Department of Neuroscience, Basic Science Building, Medical University of South Carolina, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, USA.
| | - J T McGonigal
- Department of Neuroscience, Basic Science Building, Medical University of South Carolina, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, USA
| | - K E Nimchuk
- Department of Neuroscience, Basic Science Building, Medical University of South Carolina, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, USA
| | - J T Gass
- Department of Neuroscience, Basic Science Building, Medical University of South Carolina, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, USA
- Department of Biomedical Sciences, James H. Quillen College of Medicine & VA Medical Center, PO Box 70582, Johnson City, TN, 37614, USA
| |
Collapse
|
36
|
Lguensat A, Boudjafad Z, Giorla E, Bennis M, Baunez C, Garcia R, Ba-M'hamed S. Repeated ethanol exposure following avoidance conditioning impairs avoidance extinction and modifies conditioning-associated prefrontal dendritic changes in a mouse model of post-traumatic stress disorder. Eur J Neurosci 2021; 54:7710-7732. [PMID: 34670326 DOI: 10.1111/ejn.15499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 11/28/2022]
Abstract
Treatment of post-traumatic stress disorder is complicated by the presence of alcohol use disorder comorbidity. Little is known about the underlying brain mechanisms. We have recently shown, in mice, that the post-traumatic stress disorder-like phenotype is characterised by the increase and decrease in total dendritic number and length in the prelimbic and infralimbic areas of the medial prefrontal cortex, respectively. Here, we examined whether repeated ethanol exposure would exacerbate these changes and whether this would be associated with difficulty to extinguish passive avoidance behaviour, as an indicator of treatment resistance. We also analysed whether other known trauma-associated changes, like increased or decreased corticosterone and decreased brain-derived neurotrophic factor levels, would also be exacerbated. Male mice underwent trauma exposure (1.5-mA footshock), followed, 8 days later, by a conditioned place preference training with ethanol. Tests for fear sensitization, passive avoidance, anxiety-like behaviour, extinction acquisition and relapse susceptibility were used to assess behaviour changes. Plasma corticosterone and brain-derived neurotrophic factor levels and prefrontal dendritic changes were subsequently measured. Trauma-susceptible mice exposed to ethanol acquired a strong place preference and behaved differently from those not exposed to ethanol, with delayed avoidance extinction and higher avoidance relapse vulnerability. Ethanol potentiated trauma-associated dendritic changes in the prelimbic area and suppressed trauma-associated dendritic changes in the infralimbic area. However, ethanol had no effect on trauma-induced increased corticosterone and decreased brain-derived neurotrophic factor levels. These data suggest that the modification of prefrontal trauma-related changes, due to alcohol use, can characterise, and probably support, treatment-resistant post-traumatic stress disorder.
Collapse
Affiliation(s)
- Asmae Lguensat
- Laboratoire de Pharmacologie, Neurobiologie, Anthropologie et Environnement, Université Cadi Ayyad, Marrakesh, Morocco.,Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, Aix Marseille Université, Marseille, France
| | - Zineb Boudjafad
- Laboratoire de Pharmacologie, Neurobiologie, Anthropologie et Environnement, Université Cadi Ayyad, Marrakesh, Morocco
| | - Elodie Giorla
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, Aix Marseille Université, Marseille, France
| | - Mohamed Bennis
- Laboratoire de Pharmacologie, Neurobiologie, Anthropologie et Environnement, Université Cadi Ayyad, Marrakesh, Morocco
| | - Christelle Baunez
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, Aix Marseille Université, Marseille, France
| | - René Garcia
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, Aix Marseille Université, Marseille, France.,Graduate School of Life and Health Sciences, Université Côte d'Azur, Nice, France
| | - Saadia Ba-M'hamed
- Laboratoire de Pharmacologie, Neurobiologie, Anthropologie et Environnement, Université Cadi Ayyad, Marrakesh, Morocco
| |
Collapse
|
37
|
Kipp BT, Nunes PT, Galaj E, Hitchcock B, Nasra T, Poynor KR, Heide SK, Reitz NL, Savage LM. Adolescent Ethanol Exposure Alters Cholinergic Function and Apical Dendritic Branching Within the Orbital Frontal Cortex. Neuroscience 2021; 473:52-65. [PMID: 34450212 DOI: 10.1016/j.neuroscience.2021.08.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
During adolescence, heavy binge-like ethanol consumption can lead to frontocortical structural and functional impairments. These impairments are likely driven by adolescence being a critical time point for maturation of brain regions associated with higher-order cognitive functioning. Rodent models of heavy binge-like ethanol exposure show consistent disruptions to the typical development of the prefrontal cortex (PFC). All deep cortical layers receive cholinergic projections that originate from the Nucleus basalis of Meynert (NbM) complex. These cholinergic projections are highly involved in learning, memory, and attention. Adolescent intermittent ethanol exposure (AIE) induces cholinergic dysfunction as a result of an epigenetic suppression of the genes that drive the cholinergic phenotype. The current study used a model of AIE to assess structural and functional changes to the frontal cortex and NbM following binge-like ethanol exposure in adolescence. Western blot analysis revealed long-term disruptions of the cholinergic circuit following AIE: choline acetyltransferase (ChAT) was suppressed in the NbM and vesicular acetylcholine transporter (VAChT) was suppressed in the orbitofrontal cortex (OFC). In vivo microdialysis for acetylcholine efflux during a spatial memory task determined changes in cholinergic modulation within the PFC following AIE. However, AIE spared performance on the spatial memory task and on an operant reversal task. In a second study, Golgi-Cox staining determined that AIE increased apical dendritic complexity in the OFC, with sex influencing whether the increase in branching occurred near or away from the soma. Spine density or maturity was not affected, likely compensating for a disruption in neurotransmitter function following AIE.
Collapse
Affiliation(s)
- B T Kipp
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - P T Nunes
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - E Galaj
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - B Hitchcock
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - T Nasra
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - K R Poynor
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - S K Heide
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - N L Reitz
- Department of Psychology, Binghamton University of the State University of New York, New York, USA
| | - L M Savage
- Department of Psychology, Binghamton University of the State University of New York, New York, USA.
| |
Collapse
|
38
|
Sexually dimorphic prelimbic cortex mechanisms play a role in alcohol dependence: protection by endostatin. Neuropsychopharmacology 2021; 46:1937-1949. [PMID: 34253856 PMCID: PMC8429630 DOI: 10.1038/s41386-021-01075-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 02/05/2023]
Abstract
Angiogenesis or proliferation of endothelial cells plays a role in brain microenvironment homeostasis. Previously we have shown enhanced expression of markers of angiogenesis in the medial prefrontal cortex during abstinence in an animal model of ethanol dependence induced by chronic intermittent ethanol vapor (CIE) and ethanol drinking (ED) procedure. Here we report that systemic injections of the angiogenesis inhibitor endostatin reduced relapse to drinking behavior in female CIE-ED rats without affecting relapse to drinking in male CIE-ED rats, and female and male nondependent ED rats. Endostatin did not alter relapse to sucrose drinking in both sexes. Endostatin reduced expression of platelet endothelial cell adhesion molecule-1 (PECAM-1) in all groups; however, rescued expression of tight junction protein claudin-5 in the prelimbic cortex (PLC) of female CIE-ED rats. In both sexes, CIE-ED enhanced microglial activation in the PLC and this was selectively prevented by endostatin in female CIE-ED rats. Endostatin prevented CIE-ED-induced enhanced NF-kB activity and expression and Fos expression in females and did not alter reduced Fos expression in males. Analysis of synaptic processes within the PLC revealed sexually dimorphic adaptations, with CIE-ED reducing synaptic transmission and altering synaptic plasticity in the PLC in females, and increasing synaptic transmission in males. Endostatin prevented the neuroadaptations in the PLC in females via enhancing phosphorylation of CaMKII, without affecting the neuroadaptations in males. Our multifaceted approach is the first to link PLC endothelial cell damage to the behavioral, neuroimmune, and synaptic changes associated with relapse to ethanol drinking in female subjects, and provides a new therapeutic strategy to reduce relapse in dependent subjects.
Collapse
|
39
|
Tsotsokou G, Nikolakopoulou M, Kouvelas ED, Mitsacos A. Neonatal maternal separation affects metabotropic glutamate receptor 5 expression and anxiety-related behavior of adult rats. Eur J Neurosci 2021; 54:4550-4564. [PMID: 34137089 DOI: 10.1111/ejn.15358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 01/15/2023]
Abstract
Exposure to early life stress leads to long-term neurochemical and behavioral alterations. Stress-induced psychiatric disorders, such as depression, have recently been linked to dysregulation of glutamate signaling, mainly via its postsynaptic receptors. The role of metabotropic glutamate receptor 5 (mGluR5) in stress-induced psychopathology has been the target of several studies in humans. In rodents, blockade of mGluR5 produces antidepressant-like actions, whereas mice lacking mGluR5 exhibit altered anxiety-like behaviors and learning. In this study, we used well-known rodent models of early life stress based on mother-infant separation during the first 3 weeks of life in order to examine the effects of neonatal maternal separation on mGluR5 expression and on anxiety-related behavior in adulthood. We observed that brief (15 min) neonatal maternal separation, but not prolonged (3 h), induced increases in mGluR5 mRNA and protein expression levels in medial prefrontal cortex and mGluR5 protein levels in dorsal, but not ventral, hippocampus of adult rat brain. Behavioral testing using the open-field and the elevated-plus maze tasks showed that brief maternal separations resulted in increased exploratory and decreased anxiety-related behavior, whereas prolonged maternal separations resulted in increased anxiety-related behavior in adulthood. The data indicate that the long-lasting effects of neonatal mother-offspring separation on anxiety-like behavior and mGluR5 expression depend on the duration of maternal separation and suggest that the increased mGluR5 receptors in medial prefrontal cortex and hippocampus of adult rats exposed to brief neonatal maternal separations may underlie their heightened ability to cope with stress.
Collapse
Affiliation(s)
- Giota Tsotsokou
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| | - Maria Nikolakopoulou
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| | - Elias D Kouvelas
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| | - Ada Mitsacos
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| |
Collapse
|
40
|
Avchalumov Y, Oliver RJ, Trenet W, Heyer Osorno RE, Sibley BD, Purohit DC, Contet C, Roberto M, Woodward JJ, Mandyam CD. Chronic ethanol exposure differentially alters neuronal function in the medial prefrontal cortex and dentate gyrus. Neuropharmacology 2021; 185:108438. [PMID: 33333103 PMCID: PMC7927349 DOI: 10.1016/j.neuropharm.2020.108438] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 12/06/2020] [Accepted: 12/09/2020] [Indexed: 12/28/2022]
Abstract
Alterations in the function of prefrontal cortex (PFC) and hippocampus have been implicated in underlying the relapse to alcohol seeking behaviors in humans and animal models of moderate to severe alcohol use disorders (AUD). Here we used chronic intermittent ethanol vapor exposure (CIE), 21d protracted abstinence following CIE (21d AB), and re-exposure to one vapor session during protracted abstinence (re-exposure) to evaluate the effects of chronic ethanol exposure on basal synaptic function, neuronal excitability and expression of key synaptic proteins that play a role in neuronal excitability in the medial PFC (mPFC) and dentate gyrus (DG). CIE consistently enhanced excitability of layer 2/3 pyramidal neurons in the mPFC and granule cell neurons in the DG. In the DG, this effect persisted during 21d AB. Re-exposure did not enhance excitability, suggesting resistance to vapor-induced effects. Analysis of action potential kinetics revealed that altered afterhyperpolarization, rise time and decay time constants are associated with the altered excitability during CIE, 21d AB and re-exposure. Molecular adaptations that may underlie increases in neuronal excitability under these different conditions were identified. Quantitative polymerase chain reaction of large-conductance potassium (BK) channel subunit mRNA in PFC and DG tissue homogenates did not show altered expression patterns of BK subunits. Western blotting demonstrates enhanced phosphorylation of Ca2⁺/calmodulin-dependent protein kinase II (CaMKII), and reduced phosphorylation of glutamate receptor GluN2A/2B subunits. These results suggest a novel relationship between activity of CaMKII and GluN receptors in the mPFC and DG, and neuronal excitability in these brain regions in the context of moderate to severe AUD.
Collapse
Affiliation(s)
| | | | - Wulfran Trenet
- VA San Diego Healthcare System, San Diego, CA, 92161, USA
| | | | | | | | - Candice Contet
- Departments of Molecular Medicine and Neuroscience, Scripps Research, La Jolla, CA, 92037, USA
| | - Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, Scripps Research, La Jolla, CA, 92037, USA
| | - John J Woodward
- Departments of Neuroscience and Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC, USA
| | - Chitra D Mandyam
- VA San Diego Healthcare System, San Diego, CA, 92161, USA; Departments of Molecular Medicine and Neuroscience, Scripps Research, La Jolla, CA, 92037, USA; Department of Anesthesiology, University of California San Diego, San Diego, CA, 92161, USA.
| |
Collapse
|
41
|
Somkuwar SS, Villalpando EG, Quach LW, Head BP, McKenna BS, Scadeng M, Mandyam CD. Abstinence from ethanol dependence produces concomitant cortical gray matter abnormalities, microstructural deficits and cognitive dysfunction. Eur Neuropsychopharmacol 2021; 42:22-34. [PMID: 33279357 PMCID: PMC7797163 DOI: 10.1016/j.euroneuro.2020.11.010] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/12/2020] [Accepted: 11/07/2020] [Indexed: 12/20/2022]
Abstract
Previous studies demonstrate that ethanol dependence induced by repeating cycles of chronic intermittent ethanol vapor exposure (CIE) followed by protracted abstinence (CIE-PA) produces significant alterations in oligodendrogenesis in the rodent medial prefrontal cortex (mPFC). Specifically, CIE-PA produced an unprecedented increase in premyelinating oligodendroglial progenitor cells and myelin, which have been associated with persistent elevated drinking behaviors during abstinence. The current study used neuroimaging and electron microscopy to evaluate the integrity of enhanced myelin and microstructural deficits underlying enhanced myelination in the mPFC in male rats experiencing forced abstinence for 1 day (D), 7D, 21D and 42D following seven weeks of CIE. In vivo diffusion tensor imaging (DTI) detected altered microstructural integrity in the mPFC and corpus callosum (CC). Altered integrity was characterized as reduced fractional anisotropy (FA) in the CC, and enhanced mean diffusivity (MD) in the mPFC in 7D abstinent rats. Increased MD occurred concomitantly with increases in myelin associated proteins, flayed myelin and enhanced mitochondrial stress in the mPFC in 7D abstinent rats, suggesting that the increases in myelination during abstinence was aberrant. Evaluation of cognitive performance via Pavlovian conditioning in 7D abstinent rats revealed reduced retrieval and recall of fear memories dependent on the mPFC. These findings indicate that forced abstinence from moderate to severe alcohol use disorder produces gray matter damage via myelin dysfunction in the mPFC and that these microstructural changes were associated with deficits in PFC dependent behaviors.
Collapse
Affiliation(s)
| | | | - Leon W Quach
- VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Brian P Head
- VA San Diego Healthcare System, San Diego, CA 92161, USA; Departments of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Benjamin S McKenna
- Departments of Psychiatry, University of California San Diego, San Diego, CA 92161, USA
| | - Miriam Scadeng
- Departments of Radiology, University of California San Diego, San Diego, CA 92161, USA
| | - Chitra D Mandyam
- VA San Diego Healthcare System, San Diego, CA 92161, USA; Departments of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA.
| |
Collapse
|
42
|
Cannady R, Nguyen T, Padula AE, Rinker JA, Lopez MF, Becker HC, Woodward JJ, Mulholland PJ. Interaction of chronic intermittent ethanol and repeated stress on structural and functional plasticity in the mouse medial prefrontal cortex. Neuropharmacology 2021; 182:108396. [PMID: 33181147 PMCID: PMC7942177 DOI: 10.1016/j.neuropharm.2020.108396] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/05/2020] [Accepted: 11/06/2020] [Indexed: 01/27/2023]
Abstract
Stress is a risk factor that plays a considerable role in the development and maintenance of alcohol (ethanol) abuse and relapse. Preclinical studies examining ethanol-stress interactions have demonstrated elevated ethanol drinking, cognitive deficits, and negative affective behaviors in mice. However, the neural adaptations in prefrontal cortical regions that drive these aberrant behaviors produced by ethanol-stress interactions are unknown. In this study, male C57BL/6J mice were exposed to chronic intermittent ethanol (CIE) and repeated forced swim stress (FSS). After two cycles of CIE x FSS, brain slices containing the prelimbic (PrL) and infralimbic (IfL) cortex were prepared for analysis of adaptations in dendritic spines and synaptic plasticity. In the PrL cortex, total spine density was increased in mice exposed to CIE. Immediately following induction of long-term potentiation (LTP), the fEPSP slope was increased in the PrL of CIE x FSS treated mice, indicative of a presynaptic adaptation on post-tetanic potentiation (PTP). In the IfL cortex, CIE exposure regardless of FSS experience resulted in an increase in spine density. FSS alone or when combined with CIE exposure increased PTP following LTP induction. Repeated FSS episodes increased IfL cortical paired-pulse facilitation, a second measure of presynaptic plasticity. In summary, CIE exposure resulted in structural adaptations while repeated stress exposure drove metaplastic changes in presynaptic function, demonstrating distinct morphological and functional changes in PrL and IfL cortical neurons. Thus, the structural and functional adaptations may be one mechanism underlying the development of excessive drinking and cognitive deficits associated with ethanol-stress interactions.
Collapse
Affiliation(s)
- Reginald Cannady
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA; Department of Biology, College of Science and Technology, North Carolina Agricultural & Technical State University, 1601 East Market Street, Barnes Hall 215, Greensboro, NC, 27411, USA
| | - Tiffany Nguyen
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Audrey E Padula
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Jennifer A Rinker
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Marcelo F Lopez
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Howard C Becker
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - John J Woodward
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA.
| |
Collapse
|
43
|
Muench C, Charlet K, Balderston NL, Grillon C, Heilig M, Cortes CR, Momenan R, Lohoff FW. Fear conditioning and extinction in alcohol dependence: Evidence for abnormal amygdala reactivity. Addict Biol 2021; 26:e12835. [PMID: 31702089 DOI: 10.1111/adb.12835] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/28/2019] [Accepted: 09/08/2019] [Indexed: 11/29/2022]
Abstract
Fear conditioning and extinction (FCE) are vital processes in adaptive emotion regulation and disrupted in anxiety disorders. Despite substantial comorbidity between alcohol dependence (ALC) and anxiety disorders and reports of altered negative emotion processing in ALC, neural correlates of FCE in this clinical population remain unknown. Here, we used a 2-day fear learning paradigm in 43 healthy participants and 43 individuals with ALC at the National Institutes of Health. Main outcomes of this multimodal study included structural and functional brain magnetic resonance imaging, clinical measures, as well as skin conductance responses (SCRs) to confirm differential conditioning. Successful FCE was demonstrated across participants by differential SCRs in the conditioning phase and no difference in SCRs to the conditioned stimuli in the extinction phase. The ALC group showed significantly reduced blood oxygenation level-dependent responses in the right amygdala during conditioning (Cohen's d = .89, P(FWE) = .037) and in the left amygdala during fear renewal (Cohen's d = .68, P(FWE) = .039). Right amygdala activation during conditioning was significantly correlated with ALC severity (r = .39, P(Bonferroni) = .009), depressive symptoms (r = .37, P(Bonferroni) = .015), trait anxiety (r = .41, P(Bonferroni) = .006), and perceived stress (r = .45, P(Bonferroni) = .002). Our data suggest that individuals with ALC have dysregulated fear learning, in particular, dysregulated neural activation patterns, in the amygdala. Furthermore, amygdala activation during fear conditioning was associated with ALC-related clinical measures. The FCE paradigm may be a promising tool to investigate structures involved in negative affect regulation, which might inform the development of novel treatment approaches for ALC.
Collapse
Affiliation(s)
- Christine Muench
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD USA
| | - Katrin Charlet
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD USA
| | - Nicholas L. Balderston
- Section on Neurobiology of Fear and Anxiety, National Institute of Mental Health National Institutes of Health Bethesda MD USA
| | - Christian Grillon
- Section on Neurobiology of Fear and Anxiety, National Institute of Mental Health National Institutes of Health Bethesda MD USA
| | - Markus Heilig
- Department of Clinical and Experimental Medicine Linköping University Linköping Sweden
| | - Carlos R. Cortes
- Clinical NeuroImaging Research Core, National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD USA
| | - Reza Momenan
- Clinical NeuroImaging Research Core, National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD USA
| | - Falk W. Lohoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD USA
| |
Collapse
|
44
|
Abstract
Alcohol is one of the oldest pharmacological agents used for its sedative/hypnotic effects, and alcohol abuse and alcohol use disorder (AUD) continues to be major public health issue. AUD is strongly indicated to be a brain disorder, and the molecular and cellular mechanism/s by which alcohol produces its effects in the brain are only now beginning to be understood. In the brain, synaptic plasticity or strengthening or weakening of synapses, can be enhanced or reduced by a variety of stimulation paradigms. Synaptic plasticity is thought to be responsible for important processes involved in the cellular mechanisms of learning and memory. Long-term potentiation (LTP) is a form of synaptic plasticity, and occurs via N-methyl-D-aspartate type glutamate receptor (NMDAR or GluN) dependent and independent mechanisms. In particular, NMDARs are a major target of alcohol, and are implicated in different types of learning and memory. Therefore, understanding the effect of alcohol on synaptic plasticity and transmission mediated by glutamatergic signaling is becoming important, and this will help us understand the significant contribution of the glutamatergic system in AUD. In the first part of this review, we will briefly discuss the mechanisms underlying long term synaptic plasticity in the dorsal striatum, neocortex and the hippocampus. In the second part we will discuss how alcohol (ethanol, EtOH) can modulate long term synaptic plasticity in these three brain regions, mainly from neurophysiological and electrophysiological studies. Taken together, understanding the mechanism(s) underlying alcohol induced changes in brain function may lead to the development of more effective therapeutic agents to reduce AUDs.
Collapse
Affiliation(s)
| | - 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
|
45
|
Cortez I, Rodgers SP, Kosten TA, Leasure JL. Sex and Age Effects on Neurobehavioral Toxicity Induced by Binge Alcohol. Brain Plast 2020; 6:5-25. [PMID: 33680843 PMCID: PMC7902983 DOI: 10.3233/bpl-190094] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Historically, most alcohol neurotoxicity studies were conducted in young adult males and focused on chronic intake. There has been a shift towards studying the effects of alcohol on the adolescent brain, due to alcohol consumption during this formative period disrupting the brain's developmental trajectory. Because the most typical pattern of adolescent alcohol intake is heavy episodic (binge) drinking, there has also been a shift towards the study of binge alcohol-induced neurobehavioral toxicity. It has thus become apparent that binge alcohol damages the adolescent brain and there is increasing attention to sex-dependent effects. Significant knowledge gaps remain in our understanding of the effects of binge alcohol on the female brain, however. Moreover, it is unsettling that population-level studies indicate that the prevalence of binge drinking is increasing among American women, particularly those in older age groups. Although study of adolescents has made it apparent that binge alcohol disrupts ongoing brain maturational processes, we know almost nothing about how it impacts the aging brain, as studies of its effects on the aged brain are relatively scarce, and the study of sex-dependent effects is just beginning. Given the rapidly increasing population of older Americans, it is crucial that studies address age-dependent effects of binge alcohol, and given the increase in binge drinking in older women who are at higher risk for cognitive decline relative to men, studies must encompass both sexes. Because adolescence and older age are both characterized by age-typical brain changes, and because binge drinking is the most common pattern of alcohol intake in both age groups, the knowledge that we have amassed on binge alcohol effects on the adolescent brain can inform our study of its effects on the aging brain. In this review, we therefore cover the current state of knowledge of sex and age-dependent effects of binge alcohol, as well as statistical and methodological considerations for studies aimed at addressing them.
Collapse
Affiliation(s)
- Ibdanelo Cortez
- Department of Psychology, University of Houston, Houston, TX, USA
| | | | | | - J. Leigh Leasure
- Department of Psychology, University of Houston, Houston, TX, USA
- Department of Biology & Biochemistry, University of Houston, Houston, TX, USA
| |
Collapse
|
46
|
Parekh SV, Paniccia JE, Lebonville CL, Lysle DT. Dorsal hippocampal interleukin-1 signaling mediates heroin withdrawal-enhanced fear learning. Psychopharmacology (Berl) 2020; 237:3653-3664. [PMID: 32860071 PMCID: PMC7686097 DOI: 10.1007/s00213-020-05645-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
Converging evidence suggests opioid abuse can increase the incidence and severity of post-traumatic stress disorder (PTSD) in clinical populations. Interestingly, opioid withdrawal alone can produce symptoms similar to those of PTSD. Despite this association, the neural mechanisms underlying the relationship of opioid abuse, withdrawal, and PTSD is poorly understood. Our laboratory has investigated the neurobiological underpinnings of stress-enhanced fear learning (SEFL), an animal model of PTSD-like symptoms. We have previously shown that, in SEFL, a severe footshock induces an increase in dorsal hippocampal (DH) interleukin-1β (IL-1β), and subsequent fear learning is blocked by DH IL-1 receptor antagonism (IL-1RA). Given that opioids and stress engage similar neuroimmune mechanisms, the present experiments investigate whether the same mechanisms drive heroin withdrawal to induce a PTSD-like phenotype. First, we tested the effect of a chronic escalating heroin dose and withdrawal regimen on fear learning and found it produces enhanced future fear learning. Heroin withdrawal also induces a time-dependent, region-specific increase in IL-1β and glial fibrillary acidic protein (GFAP) immunoreactivity within the dentate gyrus of the DH. IL-1β was significantly colocalized with GFAP, indicating astrocytes may be involved in increased IL-1β. Moreover, intra-DH infusions of IL-1RA 0, 24, and 48 h into heroin withdrawal prevents the development of enhanced fear learning but does not alter withdrawal-induced weight loss. Collectively, our data suggests heroin withdrawal is sufficient to produce enhanced fear learning, astrocytes may play a role in heroin withdrawal-induced IL-1β, and DH IL-1 signaling during withdrawal mediates the development of heroin withdrawal-enhanced fear learning.
Collapse
Affiliation(s)
- Shveta V. Parekh
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3270, Chapel Hill, NC 27599-3270 USA
| | - Jacqueline E. Paniccia
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3270, Chapel Hill, NC 27599-3270 USA
| | - Christina L. Lebonville
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3270, Chapel Hill, NC 27599-3270 USA
| | - Donald T. Lysle
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3270, Chapel Hill, NC 27599-3270 USA,Corresponding Author: , Telephone: +1-919-962-3088, Fax: +1-919-962-2537
| |
Collapse
|
47
|
Carlson HN, Weiner JL. The neural, behavioral, and epidemiological underpinnings of comorbid alcohol use disorder and post-traumatic stress disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:69-142. [PMID: 33648676 DOI: 10.1016/bs.irn.2020.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alcohol use disorder (AUD) and (PTSD) frequently co-occur and individuals suffering from this dual diagnosis often exhibit increased symptom severity and poorer treatment outcomes than those with only one of these diseases. Although there have been significant advances in our understanding of the neurobiological mechanisms underlying each of these disorders, the neural underpinnings of the comorbid condition remain poorly understood. This chapter summarizes recent epidemiological findings on comorbid AUD and PTSD, with a focus on vulnerable populations, the temporal relationship between these disorders, and the clinical consequences associated with the dual diagnosis. We then review animal models of the comorbid condition and emerging human and non-human animal research that is beginning to identify maladaptive neural changes common to both disorders, primarily involving functional changes in brain reward and stress networks. We end by proposing a neural framework, based on the emerging field of affective valence encoding, that may better explain the epidemiological and neural findings on AUD and PTSD.
Collapse
Affiliation(s)
- Hannah N Carlson
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jeff L Weiner
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
48
|
The infralimbic cortex and mGlu5 mediate the effects of chronic intermittent ethanol exposure on fear learning and memory. Psychopharmacology (Berl) 2020; 237:3417-3433. [PMID: 32767063 PMCID: PMC7572878 DOI: 10.1007/s00213-020-05622-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/27/2020] [Indexed: 01/19/2023]
Abstract
RATIONALE AND OBJECTIVES Alcohol use disorder (AUD) and post-traumatic stress disorder (PTSD) often occur comorbidly. While the incidence of these disorders is increasing, there is little investigation into the interacting neural mechanisms between these disorders. These studies aim to identify cognitive deficits that occur as a consequence of fear and ethanol exposure, implement a novel pharmaceutical intervention, and determine relevant underlying neurocircuitry. Additionally, due to clinical sex differences in PTSD prevalence and alcohol abuse, these studies examine the nature of this relationship in rodent models. METHODS Animals were exposed to a model of PTSD+AUD using auditory fear conditioning followed by chronic intermittent ethanol exposure (CIE). Then, rats received extinction training consisting of multiple conditioned stimulus presentations in absence of the shock. Extinction recall and context-induced freezing were measured in subsequent tests. CDPPB, a metabotropic glutamate receptor 5 (mGlu5) positive allosteric modulator, was used to treat these deficits, and region-specific effects were determined using microinjections. RESULTS These studies determined that CIE exposure led to deficits in fear extinction learning and heightened context-induced freezing while sex differences emerged in fear conditioning and extinction cue recall tests. Furthermore, using CDPPB, these studies found that enhancement of infralimbic (IfL) mGlu5 activity was able to recover CIE-induced deficits in both males and females. CONCLUSIONS These studies show that CIE induces deficits in fear-related behaviors and that enhancement of IfL glutamatergic activity can facilitate learning during extinction. Additionally, we identify novel pharmacological targets for the treatment of individuals who suffer from PTSD and AUD.
Collapse
|
49
|
Over-activation of primate subgenual cingulate cortex enhances the cardiovascular, behavioral and neural responses to threat. Nat Commun 2020; 11:5386. [PMID: 33106488 PMCID: PMC7588412 DOI: 10.1038/s41467-020-19167-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/30/2020] [Indexed: 01/10/2023] Open
Abstract
Stress-related disorders such as depression and anxiety are characterized by enhanced negative emotion and physiological dysfunction. Whilst elevated activity within area 25 of the subgenual anterior cingulate cortex (sgACC/25) has been implicated in these illnesses, it is unknown whether this over-activity is causal. By combining targeted intracerebral microinfusions with cardiovascular and behavioral monitoring in marmosets, we show that over-activation of sgACC/25 reduces vagal tone and heart rate variability, alters cortisol dynamics during stress and heightens reactivity to proximal and distal threat. 18F-FDG PET imaging shows these changes are accompanied by altered activity within a network of brain regions including the amygdala, hypothalamus and dorsolateral prefrontal cortex. Ketamine, shown to have rapid antidepressant effects, fails to reverse elevated arousal to distal threat contrary to the beneficial effects we have previously demonstrated on over-activation induced reward blunting, illustrating the symptom-specificity of its actions. Alexander et al. causally implicate over-activity in primate subgenual cingulate in affective and cardiovascular dysfunction relevant to anxiety and depression. Over-activation led to elevated activity in a stress-related network whilst decreasing activity in higher-order prefrontal cognitive regions.
Collapse
|
50
|
Shang P, Lindberg D, Starski P, Peyton L, Hong SI, Choi S, Choi DS. Chronic Alcohol Exposure Induces Aberrant Mitochondrial Morphology and Inhibits Respiratory Capacity in the Medial Prefrontal Cortex of Mice. Front Neurosci 2020; 14:561173. [PMID: 33192248 PMCID: PMC7646256 DOI: 10.3389/fnins.2020.561173] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/23/2020] [Indexed: 12/23/2022] Open
Abstract
Alcohol use disorder (AUD) is characterized as a chronic, relapsing disease with a pattern of excessive drinking despite negative consequences to an individual's life. Severe chronic alcohol use impairs the function of the medial prefrontal cortex (mPFC), which contributes to alcohol-induced cognitive and executive dysfunction. The mPFC contains more mitochondria compared to other cortical areas, which suggests mitochondrial damage may occur in AUD and trigger subsequent behavior change. Here, we identified morphological and functional changes in mitochondria in the mPFC in C57BL6/J mice after 8 h of withdrawal from chronic intermittent alcohol (CIA) exposure. Three-dimensional serial block-face scanning electron microscopy (SBFSEM) reconstruction revealed that CIA exposure elongated mPFC mitochondria and formed mitochondria-on-a-string (MOAS). Furthermore, alcohol significantly affected mitochondrial bioenergetics, including oxidative phosphorylation and electron transport, with inhibited aerobic respiration in mPFC mitochondria after CIA exposure. We also found decreased expression of fusion (mitofusin 2, Mfn2) and increased fission (mitochondrial fission 1 protein, Fis1) proteins in the mPFC of alcohol-treated mice. In sum, our study suggests that CIA exposure impairs mitochondrial dynamics and function in the mPFC.
Collapse
Affiliation(s)
- Pei Shang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Department of Neurology, First Hospital of Jilin University, Changchun, China
| | - Daniel Lindberg
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Phillip Starski
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Sa-Ik Hong
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Sun Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|