1
|
Seemiller LR, Goldberg LR, Sebastian A, Siegel SR, Praul C, Zeid D, Albert I, Beierle J, Bryant CD, Gould TJ. Alcohol and fear conditioning produce strain-specific changes in the dorsal hippocampal transcriptome of adolescent C57BL/6J and DBA/2J mice. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:2022-2034. [PMID: 39279663 DOI: 10.1111/acer.15440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/22/2024] [Accepted: 08/21/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Adolescent sensitivity to alcohol is influenced by genetic background. Data from our laboratory suggested that adolescent C57BL/6J and DBA/2J inbred mice differed in susceptibility to alcohol-induced deficits in dorsal hippocampus-dependent contextual fear learning. METHODS To investigate the biological underpinnings of this strain difference, we examined dorsal hippocampus gene expression using RNA-sequencing after alcohol or saline administration followed by Pavlovian fear conditioning across male and female C57BL/6J and DBA/2J adolescents. RESULTS Strains exhibited dramatic differences in dorsal hippocampus gene expression. Specifically, C57BL/6J and DBA/2J strains differed by 3526 transcripts in males and 2675 transcripts in females. We identified pathways likely to be involved in mediating alcohol's effects on learning, including networks associated with Chrna7, a gene encoding the nicotinic cholinergic receptor alpha 7 subunit, and Fmr1, a gene encoding the fragile X messenger ribonucleoprotein. CONCLUSIONS These findings provide insight into the mechanisms underlying strain differences in alcohol's effects on learning and suggest that different biological networks are recruited for learning based on genetics, sex, and alcohol exposure.
Collapse
Affiliation(s)
- Laurel R Seemiller
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania, USA
| | - Lisa R Goldberg
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania, USA
| | - Aswathy Sebastian
- Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania, USA
| | - Sue Rutherford Siegel
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania, USA
| | - Craig Praul
- Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania, USA
| | - Dana Zeid
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania, USA
| | - Istvan Albert
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA
| | - Jacob Beierle
- Department of Pharmacology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Camron D Bryant
- Department of Pharmaceutical Sciences, Center for Drug Discovery, Northeastern University, Boston, Massachusetts, USA
| | - Thomas J Gould
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania, USA
| |
Collapse
|
2
|
Favoretto CA, Bertagna NB, Miguel TT, Quadros IMH. The CRF/Urocortin systems as therapeutic targets for alcohol use disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 178:97-152. [PMID: 39523064 DOI: 10.1016/bs.irn.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Development and maintenance of alcohol use disorders have been proposed to recruit critical mechanisms involving Corticotropin Releasing Factor and Urocortins (CRF/Ucns). The CRF/Ucns system is comprised of a family of peptides (CRF, Ucn 1, Ucn 2, Ucn 3) which act upon two receptor subtypes, CRFR1 and CRFR2, each with different affinity profiles to the endogenous peptides and differential brain distribution. Activity of CRF/Ucn system is further modulated by CRF binding protein (CRF-BP), which regulates availability of CRF and Ucns to exert their actions. Extensive evidence in preclinical models support the involvement of CRF/Ucn targets in escalated alcohol drinking, as well as point to changes in CRF/Ucn brain function as a result of chronic alcohol exposure and/or withdrawal. It highlights the role of CRF and CRFR1-mediated signaling in conditions of excessive alcohol taking and seeking, including during various stages of withdrawal and relapse to alcohol. Besides its role in the hypothalamic-pituitary-adrenal (HPA) axis, the importance of extra-hypothalamic CRF pathways, especially in the extended amygdala, in the neurobiology of alcohol abuse and dependence is emphasized. Emerging roles for other targets of the CRF/Ucn system, such as CRF2 receptors, CRF-BP and Ucns in escalated alcohol drinking is also discussed. Finally, the limited translational value of CRF/Ucn interventions in stress-related and alcohol use disorders is discussed. So far, CRFR1 antagonists have shown little or no efficacy in human clinical trials, although a range of unexplored conditions and possibilities remain to be explored.
Collapse
Affiliation(s)
- Cristiane Aparecida Favoretto
- Molecular and Behavioral Neuroscience Laboratory, Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), SP, Brazil; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Natalia Bonetti Bertagna
- Molecular and Behavioral Neuroscience Laboratory, Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), SP, Brazil
| | | | - Isabel M H Quadros
- Psychobiology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), SP, Brazil.
| |
Collapse
|
3
|
Chan AE, Anderson JQ, Grigsby KB, Jensen BE, Ryabinin AE, Ozburn AR. Sex differences in nucleus accumbens core circuitry engaged by binge-like ethanol drinking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608144. [PMID: 39229134 PMCID: PMC11370393 DOI: 10.1101/2024.08.15.608144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Growing parity in Alcohol Use Disorder (AUD) diagnoses in men and women necessitates consideration of sex as a biological variable. In humans and rodents, the nucleus accumbens core (NAcc) regulates alcohol binge drinking, a risk factor for developing AUD. We labeled NAcc inputs with a viral retrograde tracer and quantified whole-brain c-Fos to determine the regions and NAcc inputs differentially engaged in male and female mice during binge-like ethanol drinking. We found that binge-like ethanol drinking females had 129 brain areas with greater c-Fos than males. Moreover, ethanol engaged more NAcc inputs in binge-like ethanol drinking females (as compared with males), including GABAergic and glutamatergic inputs. Relative to water controls, ethanol increased network modularity and decreased connectivity in both sexes and did so more dramatically in males. These results demonstrate that early-stage binge-like ethanol drinking engages brain regions and NAcc-inputs and alters network dynamics in a sex-specific manner.
Collapse
Affiliation(s)
- Amy E Chan
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Justin Q Anderson
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Kolter B Grigsby
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Bryan E Jensen
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Andrey E Ryabinin
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
| | - Angela R Ozburn
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| |
Collapse
|
4
|
Maddern XJ, Ursich LT, Bailey G, Pearl A, Anversa RG, Lawrence AJ, Walker LC. Sex Differences in Alcohol Use: Is It All About Hormones? Endocrinology 2024; 165:bqae088. [PMID: 39018449 DOI: 10.1210/endocr/bqae088] [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: 04/02/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/19/2024]
Abstract
Risky alcohol use and alcohol use disorders (AUD) are a rising problem in women, yet a major disparity in our understanding of what drives alcohol consumption in women remains. Historically biomedical research has focused on male subjects; however, recent increases in reporting of females, have highlighted major differences between the sexes. Here we review the current literature of the effect of gonadal steroid hormones (estrogens, androgens, and progestins), neurosteriods, and neurobiological factors on alcohol use in clinical and preclinical studies of both sexes. Further, we briefly discuss how fundamental sex differences in genetics, metabolism, neuroimmune, and stress responses may influence sex differences in alcohol intake. Comparing the sexes could aid in the discovery of novel therapeutics to treat AUD, and implementation of current treatment options in women.
Collapse
Affiliation(s)
- Xavier J Maddern
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lauren T Ursich
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Grace Bailey
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Amy Pearl
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
| | - Roberta G Anversa
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| |
Collapse
|
5
|
Brocato ER, Easter R, Morgan A, Kakani M, Lee G, Wolstenholme JT. Adolescent binge ethanol impacts H3K9me3-occupancy at synaptic genes and the regulation of oligodendrocyte development. Front Mol Neurosci 2024; 17:1389100. [PMID: 38840776 PMCID: PMC11150558 DOI: 10.3389/fnmol.2024.1389100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction Binge drinking in adolescence can disrupt myelination and cause brain structural changes that persist into adulthood. Alcohol consumption at a younger age increases the susceptibility of these changes. Animal models to understand ethanol's actions on myelin and white matter show that adolescent binge ethanol can alter the developmental trajectory of oligodendrocytes, myelin structure, and myelin fiber density. Oligodendrocyte differentiation is epigenetically regulated by H3K9 trimethylation (H3K9me3). Prior studies have shown that adolescent binge ethanol dysregulates H3K9 methylation and decreases H3K9-related gene expression in the PFC. Methods Here, we assessed ethanol-induced changes to H3K9me3 occupancy at genomic loci in the developing adolescent PFC. We further assessed ethanol-induced changes at the transcription level with qPCR time course approaches in oligodendrocyte-enriched cells to assess changes in oligodendrocyte progenitor and oligodendrocytes specifically. Results Adolescent binge ethanol altered H3K9me3 regulation of synaptic-related genes and genes specific for glutamate and potassium channels in a sex-specific manner. In PFC tissue, we found an early change in gene expression in transcription factors associated with oligodendrocyte differentiation that may lead to the later significant decrease in myelin-related gene expression. This effect appeared stronger in males. Conclusion Further exploration in oligodendrocyte cell enrichment time course and dose response studies could suggest lasting dysregulation of oligodendrocyte maturation at the transcriptional level. Overall, these studies suggest that binge ethanol may impede oligodendrocyte differentiation required for ongoing myelin development in the PFC by altering H3K9me3 occupancy at synaptic-related genes. We identify potential genes that may be contributing to adolescent binge ethanol-related myelin loss.
Collapse
Affiliation(s)
- Emily R. Brocato
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
| | - Rachel Easter
- Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Alanna Morgan
- Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Meenakshi Kakani
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
| | - Grace Lee
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennifer T. Wolstenholme
- Pharmacology and Toxicology Department, Virginia Commonwealth University, Richmond, VA, United States
- Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
6
|
Finn DA, Clark CD, Ryabinin AE. Traumatic stress-enhanced alcohol drinking: Sex differences and animal model perspectives. CURRENT ADDICTION REPORTS 2024; 11:327-341. [PMID: 38915732 PMCID: PMC11196023 DOI: 10.1007/s40429-023-00540-0] [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] [Accepted: 12/27/2023] [Indexed: 06/26/2024]
Abstract
Purpose of review Stress is associated with alcohol drinking, and epidemiological studies document the comorbidity of alcohol use disorder (AUD) and post-traumatic stress disorder (PTSD), with higher comorbid prevalence in females than in males. The aim of this paper is to highlight information related to sex differences in stress-enhanced alcohol drinking from clinical studies and from preclinical studies utilizing an animal model of traumatic stress. Recent findings Stress is associated with alcohol drinking and relapse in males and females, but there are sex differences in the alcohol-related adaptation of stress pathways and in the association of different prefrontal regions with stress-induced anxiety. The predator stress model of traumatic stress produced enhanced alcohol drinking in a subgroup of stress-sensitive male and female animals, which could be associated with sex and subgroup differences in stress axis responsivity, behavioral responses to predator odors, and epigenetic mechanisms engaged by traumatic experiences. Summary While additional studies in females are necessary, existing clinical and preclinical evidence suggests that biological mechanisms underlying stress-enhanced drinking likely differ between males and females. Thus, effective treatment strategies may differ between the sexes.
Collapse
Affiliation(s)
- Deborah A. Finn
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Department of Research, VA Portland Health Care System, Portland, OR, United States
| | - Crystal D. Clark
- Department of Research, VA Portland Health Care System, Portland, OR, United States
| | - Andrey E. Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
7
|
North KC, Mysiewicz SC, Bukiya AN, Dopico AM. Dual-color miniscope imaging of microvessels and neuronal activity in the hippocampus CA1 region of freely moving mice following alcohol administration. Am J Physiol Regul Integr Comp Physiol 2023; 325:R769-R781. [PMID: 37867475 PMCID: PMC11178301 DOI: 10.1152/ajpregu.00044.2023] [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: 02/16/2023] [Revised: 09/08/2023] [Accepted: 10/12/2023] [Indexed: 10/24/2023]
Abstract
Moderate-to-heavy episodic ("binge") drinking is the most common form of alcohol consumption in the United States. Alcohol at binge drinking concentrations reduces brain artery diameter in vivo and in vitro in many species including rats, mice, and humans. Despite the critical role played by brain vessels in maintaining neuronal function, there is a shortage of methodologies to simultaneously assess neuron and blood vessel function in deep brain regions. Here, we investigate cerebrovascular responses to ethanol by choosing a deep brain region that is implicated in alcohol disruption of brain function, the hippocampal CA1, and describe the process for obtaining simultaneous imaging of pyramidal neuron activity and diameter of nearby microvessels in freely moving mice via a dual-color miniscope. Recordings of neurovascular events were performed upon intraperitoneal injection of saline versus 3 g/kg ethanol in the same mouse. In male mice, ethanol mildly increased the amplitude of calcium signals while robustly decreasing their frequency. Simultaneously, ethanol decreased microvessel diameter. In females, ethanol did not change the amplitude or frequency of calcium signals from CA1 neurons but decreased microvessel diameter. A linear regression of ethanol-induced reduction in number of active neurons and microvessel constriction revealed a positive correlation (R = 0.981) in females. Together, these data demonstrate the feasibility of simultaneously evaluating neuronal and vascular components of alcohol actions in a deep brain area in freely moving mice, as well as the sexual dimorphism of hippocampal neurovascular responses to alcohol.
Collapse
Affiliation(s)
- Kelsey C North
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Steven C Mysiewicz
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| |
Collapse
|
8
|
Finn DA. Stress and gonadal steroid influences on alcohol drinking and withdrawal, with focus on animal models in females. Front Neuroendocrinol 2023; 71:101094. [PMID: 37558184 PMCID: PMC10840953 DOI: 10.1016/j.yfrne.2023.101094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/06/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023]
Abstract
Sexually dimorphic effects of alcohol, following binge drinking, chronic intoxication, and withdrawal, are documented at the level of the transcriptome and in behavioral and physiological responses. The purpose of the current review is to update and to expand upon contributions of the endocrine system to alcohol drinking and withdrawal in females, with a focus on animal models. Steroids important in the hypothalamic-pituitary-gonadal and hypothalamic-pituitary-adrenal axes, the reciprocal interactions between these axes, the effects of chronic alcohol use on steroid levels, and the genomic and rapid membrane-associated effects of steroids and neurosteroids in models of alcohol drinking and withdrawal are described. Importantly, comparison between males and females highlight some divergent effects of sex- and stress-steroids on alcohol drinking- and withdrawal-related behaviors, and the distinct differences in response emphasize the importance of considering sex in the development of novel pharmacotherapies for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Deborah A Finn
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States; Department of Research, VA Portland Health Care System, Portland, OR, United States.
| |
Collapse
|
9
|
Szumlinski KK, Herbert JN, Mejia Espinoza B, Madory LE, Scudder SL. Alcohol-drinking during later life by C57BL/6J mice induces sex- and age-dependent changes in hippocampal and prefrontal cortex expression of glutamate receptors and neuropathology markers. ADDICTION NEUROSCIENCE 2023; 7:100099. [PMID: 37396410 PMCID: PMC10310297 DOI: 10.1016/j.addicn.2023.100099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Heavy drinking can induce early-onset dementia and increase the likelihood of the progression and severity of Alzheimer's Disease and related dementias (ADRD). Recently, we showed that alcohol-drinking by mature adult C57BL/6J mice induces more signs of cognitive impairment in females versus males without worsening age-related cognitive decline in aged mice. Here, we immunoblotted for glutamate receptors and protein markers of ADRD-related neuropathology within the hippocampus and prefrontal cortex (PFC) of these mice after three weeks of alcohol withdrawal to determine protein correlates of alcohol-induced cognitive decline. Irrespective of alcohol history, age-related changes in protein expression included a male-specific decline in hippocampal glutamate receptors and an increase in the expression of a beta-site amyloid precursor protein cleaving enzyme (BACE) isoform in the PFC as well as a sex-independent increase in hippocampal amyloid precursor protein. Alcohol-drinking was associated with altered expression of glutamate receptors in the hippocampus in a sex-dependent manner, while all glutamate receptor proteins exhibited significant alcohol-related increases in the PFC of both sexes. Expression of BACE isoforms and phosphorylated tau varied in the PFC and hippocampus based on age, sex, and drinking history. The results of this study indicate that withdrawal from a history of alcohol-drinking during later life induces sex- and age-selective effects on glutamate receptor expression and protein markers of ADRD-related neuropathology within the hippocampus and PFC of potential relevance to the etiology, treatment and prevention of alcohol-induced dementia and Alzheimer's Disease.
Collapse
Affiliation(s)
- Karen K. Szumlinski
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106-9625, USA
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106-9625, USA
| | - Jessica N. Herbert
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
| | - Brenda Mejia Espinoza
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
| | - Lauren E. Madory
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
| | - Samantha L. Scudder
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
- Department of Psychology, California State University Dominguez Hills, Carson, CA 90747, USA
| |
Collapse
|
10
|
Cruz B, Borgonetti V, Bajo M, Roberto M. Sex-dependent factors of alcohol and neuroimmune mechanisms. Neurobiol Stress 2023; 26:100562. [PMID: 37601537 PMCID: PMC10432974 DOI: 10.1016/j.ynstr.2023.100562] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/25/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023] Open
Abstract
Excessive alcohol use disrupts neuroimmune signaling across various cell types, including neurons, microglia, and astrocytes. The present review focuses on recent, albeit limited, evidence of sex differences in biological factors that mediate neuroimmune responses to alcohol and underlying neuroimmune systems that may influence alcohol drinking behaviors. Females are more vulnerable than males to the neurotoxic and negative consequences of chronic alcohol drinking, reflected by elevations of pro-inflammatory cytokines and inflammatory mediators. Differences in cytokine, microglial, astrocytic, genomic, and transcriptomic evidence suggest females are more reactive than males to neuroinflammatory changes after chronic alcohol exposure. The growing body of evidence supports that innate immune factors modulate synaptic transmission, providing a mechanistic framework to examine sex differences across neurocircuitry. Targeting neuroimmune signaling may be a viable strategy for treating AUD, but more research is needed to understand sex-specific differences in alcohol drinking and neuroimmune mechanisms.
Collapse
Affiliation(s)
- Bryan Cruz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Vittoria Borgonetti
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| |
Collapse
|
11
|
Cheng Y, Dempsey RE, Roodsari SK, Shuboni-Mulligan DD, George O, Sanford LD, Guo ML. Cocaine Regulates NLRP3 Inflammasome Activity and CRF Signaling in a Region- and Sex-Dependent Manner in Rat Brain. Biomedicines 2023; 11:1800. [PMID: 37509440 PMCID: PMC10376186 DOI: 10.3390/biomedicines11071800] [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: 05/03/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Cocaine, one of the most abused drugs worldwide, is capable of activating microglia in vitro and in vivo. Several neuroimmune pathways have been suggested to play roles in cocaine-mediated microglial activation. Previous work showed that cocaine activates microglia in a region-specific manner in the brains of self-administered mice. To further characterize the effects of cocaine on microglia and neuroimmune signaling in vivo, we utilized the brains from both sexes of outbred rats with cocaine self-administration to explore the activation status of microglia, NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activity, corticotropin-releasing factor (CRF) signaling, and NF-κB levels in the striatum and hippocampus (HP). Age-matched rats of the same sex (drug naïve) served as controls. Our results showed that cocaine increased neuroinflammation in the striatum and HP of both sexes with a relatively higher increases in male brains. In the striatum, cocaine upregulated NLRP3 inflammasome activity and CRF levels in males but not in females. In contrast, cocaine increased NLRP3 inflammasome activity in the HP of females but not in males, and no effects on CRF signaling were observed in this region of either sex. Interestingly, cocaine increased NF-κB levels in the striatum and HP with no sex difference. Taken together, our results provide evidence that cocaine can exert region- and sex-specific differences in neuroimmune signaling in the brain. Targeting neuroimmune signaling has been suggested as possible treatment for cocaine use disorders (CUDs). Our current results indicate that sex should be taken into consideration when determining the efficacy of these new therapeutic approaches.
Collapse
Affiliation(s)
- Yan Cheng
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Rachael Elizabeth Dempsey
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Soheil Kazemi Roodsari
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Dorela D Shuboni-Mulligan
- Sleep Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Olivier George
- Department of Psychiatry, School of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Larry D Sanford
- Sleep Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Ming-Lei Guo
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| |
Collapse
|
12
|
Brocato ER, Wolstenholme JT. Adolescent binge ethanol impacts H3K36me3 regulation of synaptic genes. Front Mol Neurosci 2023; 16:1082104. [PMID: 36937047 PMCID: PMC10020663 DOI: 10.3389/fnmol.2023.1082104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Adolescence is marked in part by the ongoing development of the prefrontal cortex (PFC). Binge ethanol use during this critical stage in neurodevelopment induces significant structural changes to the PFC, as well as cognitive and behavioral deficits that can last into adulthood. Previous studies showed that adolescent binge ethanol causes lasting deficits in working memory, decreases in the expression of chromatin remodeling genes responsible for the methylation of histone 3 lysine 36 (H3K36), and global decreases in H3K36 in the PFC. H3K36me3 is present within the coding region of actively-transcribed genes, and safeguards against aberrant, cryptic transcription by RNA Polymerase II. We hypothesize that altered methylation of H3K36 could play a role in adolescent binge ethanol-induced memory deficits. To investigate this at the molecular level, ethanol (4 g/kg, i.g.) or water was administered intermittently to adolescent mice. RNA-and ChIP-sequencing were then performed within the same tissue to determine gene expression changes and identify genes and loci where H3K36me3 was disrupted by ethanol. We further assessed ethanol-induced changes at the transcription level with differential exon-use and cryptic transcription analysis - a hallmark of decreased H3K36me3. Here, we found ethanol-induced changes to the gene expression and H3K36me3-regulation of synaptic-related genes in all our analyses. Notably, H3K36me3 was differentially trimethylated between ethanol and control conditions at synaptic-related genes, and Snap25 and Cplx1 showed evidence of cryptic transcription in males and females treated with ethanol during adolescence. Our results provide preliminary evidence that ethanol-induced changes to H3K36me3 during adolescent neurodevelopment may be linked to synaptic dysregulation at the transcriptional level, which may explain the reported ethanol-induced changes to PFC synaptic function.
Collapse
Affiliation(s)
- Emily R. Brocato
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennifer T. Wolstenholme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
13
|
Plasil SL, Collins VJ, Baratta AM, Farris SP, Homanics GE. Hippocampal ceRNA networks from chronic intermittent ethanol vapor-exposed male mice and functional analysis of top-ranked lncRNA genes for ethanol drinking phenotypes. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10831. [PMID: 36908580 PMCID: PMC10004261 DOI: 10.3389/adar.2022.10831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The molecular mechanisms regulating the development and progression of alcohol use disorder (AUD) are largely unknown. While noncoding RNAs have previously been implicated as playing key roles in AUD, long-noncoding RNA (lncRNA) remains understudied in relation to AUD. In this study, we first identified ethanol-responsive lncRNAs in the mouse hippocampus that are transcriptional network hub genes. Microarray analysis of lncRNA, miRNA, circular RNA, and protein coding gene expression in the hippocampus from chronic intermittent ethanol vapor- or air- (control) exposed mice was used to identify ethanol-responsive competing endogenous RNA (ceRNA) networks. Highly interconnected lncRNAs (genes that had the strongest overall correlation to all other dysregulated genes identified) were ranked. The top four lncRNAs were novel, previously uncharacterized genes named Gm42575, 4930413E15Rik, Gm15767, and Gm33447, hereafter referred to as Pitt1, Pitt2, Pitt3, and Pitt4, respectively. We subsequently tested the hypothesis that CRISPR/Cas9 mutagenesis of the putative promoter and first exon of these lncRNAs in C57BL/6J mice would alter ethanol drinking behavior. The Drinking in the Dark (DID) assay was used to examine binge-like drinking behavior, and the Every-Other-Day Two-Bottle Choice (EOD-2BC) assay was used to examine intermittent ethanol consumption and preference. No significant differences between control and mutant mice were observed in the DID assay. Female-specific reductions in ethanol consumption were observed in the EOD-2BC assay for Pitt1, Pitt3, and Pitt4 mutant mice compared to controls. Male-specific alterations in ethanol preference were observed for Pitt1 and Pitt2. Female-specific increases in ethanol preference were observed for Pitt3 and Pitt4. Total fluid consumption was reduced in Pitt1 and Pitt2 mutants at 15% v/v ethanol and in Pitt3 and Pitt4 at 20% v/v ethanol in females only. We conclude that all lncRNAs targeted altered ethanol drinking behavior, and that lncRNAs Pitt1, Pitt3, and Pitt4 influenced ethanol consumption in a sex-specific manner. Further research is necessary to elucidate the biological mechanisms for these effects. These findings add to the literature implicating noncoding RNAs in AUD and suggest lncRNAs also play an important regulatory role in the disease.
Collapse
Affiliation(s)
- SL Plasil
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - VJ Collins
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - AM Baratta
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - SP Farris
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - GE Homanics
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Borrego MB, Chan AE, Ozburn AR. Regulation of alcohol drinking by ventral striatum and extended amygdala circuitry. Neuropharmacology 2022; 212:109074. [PMID: 35487273 PMCID: PMC9677601 DOI: 10.1016/j.neuropharm.2022.109074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/24/2022] [Accepted: 04/20/2022] [Indexed: 02/07/2023]
Abstract
Alcohol use disorder is a complex psychiatric disorder that can be modeled in rodents using a number of drinking paradigms. Drinking-in-the-dark (DID) is widely used to model the binge/intoxication stage of addiction, and chronic intermittent ethanol vapor procedures (CIE) are used to induce dependence and model withdrawal/negative affect induced escalation of drinking. We discuss experiments showing the ventral striatum (vStr) and extended amygdala (EA) are engaged in response to ethanol in rodents through c-Fos/Fos immunoreactivity studies. We also discuss experiments in rodents that span a wide variety of techniques where the function of vStr and EA structures are changed following DID or CIE, and the role of neurotransmitter and neuropeptide systems studies in these ethanol-related outcomes. We note where signaling systems converge across regions and paradigms and where there are still gaps in the literature. Dynorphin/κ-opioid receptor (KOR) signaling, as well as corticotropin releasing factor (CRF)/CRF receptor signaling were found to be important regulators of drinking behaviors across brain regions and drinking paradigms. Future research will require that females and a variety of rodent strains are used in preclinical experiments in order to strengthen the generalizability of findings and improve the likelihood of success for testing potential therapeutics in human laboratory studies.
Collapse
Affiliation(s)
- Marissa B Borrego
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Amy E Chan
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Angela R Ozburn
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA.
| |
Collapse
|
15
|
Mineur YS, Garcia-Rivas V, Thomas MA, Soares AR, McKee SA, Picciotto MR. Sex differences in stress-induced alcohol intake: a review of preclinical studies focused on amygdala and inflammatory pathways. Psychopharmacology (Berl) 2022; 239:2041-2061. [PMID: 35359158 PMCID: PMC9704113 DOI: 10.1007/s00213-022-06120-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/14/2022] [Indexed: 02/06/2023]
Abstract
Clinical studies suggest that women are more likely than men to relapse to alcohol drinking in response to stress; however, the mechanisms underlying this sex difference are not well understood. A number of preclinical behavioral models have been used to study stress-induced alcohol intake. Here, we review paradigms used to study effects of stress on alcohol intake in rodents, focusing on findings relevant to sex differences. To date, studies of sex differences in stress-induced alcohol drinking have been somewhat limited; however, there is evidence that amygdala-centered circuits contribute to effects of stress on alcohol seeking. In addition, we present an overview of inflammatory pathways leading to microglial activation that may contribute to alcohol-dependent behaviors. We propose that sex differences in neuronal function and inflammatory signaling in circuits centered on the amygdala are involved in sex-dependent effects on stress-induced alcohol seeking and suggest that this is an important area for future studies.
Collapse
Affiliation(s)
- Yann S Mineur
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Vernon Garcia-Rivas
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Merrilee A Thomas
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Alexa R Soares
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA
| | - Sherry A McKee
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA.
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA.
| |
Collapse
|
16
|
Ferguson LB, Roberts AJ, Mayfield RD, Messing RO. Blood and brain gene expression signatures of chronic intermittent ethanol consumption in mice. PLoS Comput Biol 2022; 18:e1009800. [PMID: 35176017 PMCID: PMC8853518 DOI: 10.1371/journal.pcbi.1009800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 01/03/2022] [Indexed: 02/03/2023] Open
Abstract
Alcohol Use Disorder (AUD) is a chronic, relapsing syndrome diagnosed by a heterogeneous set of behavioral signs and symptoms. There are no laboratory tests that provide direct objective evidence for diagnosis. Microarray and RNA-Seq technologies enable genome-wide transcriptome profiling at low costs and provide an opportunity to identify biomarkers to facilitate diagnosis, prognosis, and treatment of patients. However, access to brain tissue in living patients is not possible. Blood contains cellular and extracellular RNAs that provide disease-relevant information for some brain diseases. We hypothesized that blood gene expression profiles can be used to diagnose AUD. We profiled brain (prefrontal cortex, amygdala, and hypothalamus) and blood gene expression levels in C57BL/6J mice using RNA-seq one week after chronic intermittent ethanol (CIE) exposure, a mouse model of alcohol dependence. We found a high degree of preservation (rho range: [0.50, 0.67]) between blood and brain transcript levels. There was small overlap between blood and brain DEGs, and considerable overlap of gene networks perturbed after CIE related to cell-cell signaling (e.g., GABA and glutamate receptor signaling), immune responses (e.g., antigen presentation), and protein processing / mitochondrial functioning (e.g., ubiquitination, oxidative phosphorylation). Blood gene expression data were used to train classifiers (logistic regression, random forest, and partial least squares discriminant analysis), which were highly accurate at predicting alcohol dependence status (maximum AUC: 90.1%). These results suggest that gene expression profiles from peripheral blood samples contain a biological signature of alcohol dependence that can discriminate between CIE and Air subjects.
Collapse
Affiliation(s)
- Laura B. Ferguson
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, United States of America
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, Texas, United States of America
- Department of Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
| | - Amanda J. Roberts
- Animal Models Core Facility, The Scripps Research Institute, San Diego, California, United States of America
| | - R. Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, United States of America
- Department of Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
| | - Robert O. Messing
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, United States of America
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, Texas, United States of America
- Department of Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
| |
Collapse
|
17
|
Agoglia AE, Zhu M, Quadir SG, Bluitt MN, Douglass E, Hanback T, Tella J, Ying R, Hodge CW, Herman MA. Sex-specific plasticity in CRF regulation of inhibitory control in central amygdala CRF1 neurons after chronic voluntary alcohol drinking. Addict Biol 2022; 27:e13067. [PMID: 34075665 PMCID: PMC8636550 DOI: 10.1111/adb.13067] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/01/2021] [Accepted: 05/14/2021] [Indexed: 01/03/2023]
Abstract
Despite strong preclinical evidence for the ability of corticotropin releasing factor 1 (CRF1) antagonists to regulate alcohol consumption, clinical trials have not yet demonstrated therapeutic effects of these compounds in alcohol use disorder (AUD) patients. Several confounding factors may limit the translation of preclinical CRF1 research to patients, including reliance on experimenter-administered alcohol instead of voluntary consumption, a preponderance of evidence collected in male subjects only and an inability to assess the effects of alcohol on specific brain circuits. A population of particular interest is the CRF1-containing neurons of the central amygdala (CeA). CRF1 CeA neurons are sensitive to ethanol, but the effects of alcohol drinking on CRF signalling within this population are unknown. In the present study, we assessed the effects of voluntary alcohol drinking on inhibitory control of CRF1+ CeA neurons from male and female CRF1:GFP mice using ex vivo electrophysiology and determined the contributions of CRF1 signalling to inhibitory control and voluntary alcohol drinking. Chronic alcohol drinking produced neuroadaptations in CRF1+ neurons that increased the sensitivity of GABAA receptor-mediated sIPSCs to the acute effects of alcohol, CRF and the CRF1 antagonist R121919, but these adaptations were more pronounced in male versus female mice. The CRF1 antagonist CP-154,526 reduced voluntary alcohol drinking in both sexes and abolished sex differences in alcohol drinking. The lack of alcohol-induced adaptation in the female CRF1 system may be related to the elevated alcohol intake exhibited by female mice and could contribute to the ineffectiveness of CRF1 antagonists in female AUD patients.
Collapse
Affiliation(s)
- AE Agoglia
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - M Zhu
- Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - SG Quadir
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - MN Bluitt
- Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - E Douglass
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - T Hanback
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - J Tella
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - R Ying
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - CW Hodge
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - MA Herman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
18
|
Joo YH, Kim JH, Kim HK, Son YD, Cumming P, Kim JH. Functional Analysis of Brain Imaging Suggests Changes in the Availability of mGluR5 and Altered Connectivity in the Cerebral Cortex of Long-Term Abstaining Males with Alcohol Dependence: A Preliminary Study. Life (Basel) 2021; 11:life11060506. [PMID: 34070900 PMCID: PMC8228527 DOI: 10.3390/life11060506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/23/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022] Open
Abstract
Direct in vivo evidence of altered metabotropic glutamate receptor-5 (mGluR5) availability in alcohol-related disorders is lacking. We performed [11C]ABP688 positron emission tomography (PET) and resting-state functional magnetic resonance imaging (rs-fMRI) in prolonged abstinent subjects with alcohol dependence to examine alterations of mGluR5 availability, and to investigate their functional significance relating to neural systems-level changes. Twelve prolonged abstinent male subjects with alcohol dependence (median abstinence duration: six months) and ten healthy male controls underwent [11C]ABP688 PET imaging and 3-Tesla MRI. For mGluR5 availability, binding potential (BPND) was calculated using the simplified reference tissue model with cerebellar gray matter as the reference region. The initial region-of-interest (ROI)-based analysis yielded no significant group differences in mGluR5 availability. The voxel-based analysis revealed significantly lower [11C]ABP688 BPND in the middle temporal and inferior parietal cortices, and higher BPND in the superior temporal cortex in the alcohol dependence group compared with controls. Functional connectivity analysis of the rs-fMRI data employed seed regions identified from the quantitative [11C]ABP688 PET analysis, which revealed significantly altered functional connectivity from the inferior parietal cortex seed to the occipital pole and dorsal visual cortex in the alcohol dependence group compared with the control group. To our knowledge, this is the first report on the combined analysis of mGluR5 PET imaging and rs-fMRI in subjects with alcohol dependence. These preliminary results suggest the possibility of region-specific alterations of mGluR5 availability in vivo and related functional connectivity perturbations in prolonged abstinent subjects.
Collapse
Affiliation(s)
- Yo-Han Joo
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (J.-H.K.); (H.-K.K.)
| | - Jeong-Hee Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (J.-H.K.); (H.-K.K.)
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon 21936, Korea
| | - Hang-Keun Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (J.-H.K.); (H.-K.K.)
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon 21936, Korea
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
| | - Young-Don Son
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (J.-H.K.); (H.-K.K.)
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon 21936, Korea
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
- Correspondence: (Y.-D.S.); or (J.-H.K.); Tel.: +82-32-820-4416 (Y.-D.S.); +82-32-460-2696 (J.-H.K.)
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, University of Bern, CH-3010 Bern, Switzerland;
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Jong-Hoon Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (J.-H.K.); (H.-K.K.)
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
- Gil Medical Center, Department of Psychiatry, Gachon University College of Medicine, Gachon University, Incheon 21565, Korea
- Correspondence: (Y.-D.S.); or (J.-H.K.); Tel.: +82-32-820-4416 (Y.-D.S.); +82-32-460-2696 (J.-H.K.)
| |
Collapse
|
19
|
Sex Differences in the Brain Transcriptome Related to Alcohol Effects and Alcohol Use Disorder. Biol Psychiatry 2021; 91:43-52. [PMID: 34274109 PMCID: PMC8558111 DOI: 10.1016/j.biopsych.2021.04.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/05/2021] [Accepted: 04/26/2021] [Indexed: 01/03/2023]
Abstract
There is compelling evidence that sex and gender have crucial roles in excessive alcohol (ethanol) consumption. Here, we review some of the data from the perspective of brain transcriptional differences between males and females, focusing on rodent animal models. A key emerging transcriptional feature is the role of neuroimmune processes. Microglia are the resident neuroimmune cells in the brain and exhibit substantial functional differences between males and females. Selective breeding for binge ethanol consumption and the impacts of chronic ethanol consumption and withdrawal from chronic ethanol exposure all demonstrate sex-dependent neuroimmune signatures. A focus is on resolving sex-dependent differences in transcriptional responses to ethanol at the neurocircuitry level. Sex-dependent transcriptional differences are found in the extended amygdala and the nucleus accumbens. Telescoping of ethanol consumption is found in some, but not all, studies to be more prevalent in females. Recent transcriptional studies suggest that some sex differences may be due to female-dependent remodeling of the primary cilium. An interesting theme appears to be developing: at least from the animal model perspective, even when males and females are phenotypically similar, they differ significantly at the level of the transcriptome.
Collapse
|
20
|
Huang G, Thompson SL, Taylor JR. MPEP Lowers Binge Drinking in Male and Female C57BL/6 Mice: Relationship with mGlu5/Homer2/Erk2 Signaling. Alcohol Clin Exp Res 2021; 45:732-742. [PMID: 33587295 PMCID: PMC8076072 DOI: 10.1111/acer.14576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/10/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Metabotropic glutamate receptor 5 (mGlu5) plays an important role in excessive alcohol use and the mGlu5/Homer2/Erk2 signaling pathway has been implicated in binge drinking. The mGlu5 negative allosteric modulator (NAM) 2-methyl-6-(phenylethynyl)pyridine hydrochloride (MPEP) has been shown to reduce binge drinking in male mice, but less is known about its effect on female mice. Here, we sought to determine whether sex differences exists in the effects of MPEP on binge drinking and whether they relate to changes in the MPEP mGlu5/Homer2/Erk2 signaling. METHODS We measured the dose-response effect of MPEP on alcohol consumption in male and female mice using the Drinking in the Dark (DID) paradigm to assess potential sex differences. To rule out possible confounds of MPEP on locomotion, we measured the effects of MPEP on locomotor activity and drinking simultaneously during DID. Lastly, to test whether MPEP-induced changes in alcohol consumption were related to changes in Homer2 or Erk2 expression, we performed qPCR using brain tissue acquired from mice that had undergone 7 days of DID. RESULTS 30 mg/kg MPEP reduced binge alcohol consumption across female and male mice, with no sex differences in the dose-response relationship. Locomotor activity did not mediate the effects of MPEP on alcohol intake, but activity correlated with alcohol intake independent of MPEP. MPEP did not change the expression of Homer2 and Erk2 mRNA in the bed nucleus of the stria terminalis (BNST) or nucleus accumbens in mice whose drinking was reduced by MPEP, relative to saline. There was a positive relationship between alcohol intake and Homer2 expression in the BNST. CONCLUSIONS MPEP reduced alcohol consumption during DID in male and female C57BL/6 mice but did not change Homer2/Erk2 expression. Locomotor activity did not mediate the effects of MPEP on alcohol intake, though it correlated with alcohol intake. Alcohol intake during DID predicted BNST Homer2 expression. These data provide support for the regulation of alcohol consumption by mGlu5 across sexes.
Collapse
Affiliation(s)
- Gan Huang
- Department of Psychiatry, Division of Molecular Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Summer L. Thompson
- Department of Psychiatry, Division of Molecular Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Jane R. Taylor
- Department of Psychiatry, Division of Molecular Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychology, Yale University Graduate School of Arts and Sciences, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
21
|
Moench KM, Logrip ML. Housing Condition Differentially Impacts Escalation of Alcohol Intake, Relapse-Like Drinking, Anxiety-Like Behavior, and Stress History Effects by Sex. Alcohol Clin Exp Res 2021; 45:480-489. [PMID: 33351976 PMCID: PMC7890766 DOI: 10.1111/acer.14540] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/16/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Stress triggers alcohol use and relapse to drinking, with different effects by sex. Women are more susceptible to stress-related alcohol misuse, and most stressors in rodents produce sexually divergent effects. Female rodents are particularly sensitive to the stress produced by solitary housing, yet the impact of housing conditions on the establishment, escalation, and post-abstinence potentiation of intermittent access alcohol drinking in male and female rats, and the interaction of these factors with stress history are not well described. METHODS Male (n = 62) and female (n = 64) Wistar rats were housed individually or in pairs separated by a perforated divider. Rats were exposed to light-cued footshock stress (stress history), or cues alone (control), once daily for 3 days, followed by 8 weeks' drinking under intermittent access to a 2-bottle choice (IA2BC), with 20% alcohol (v/v in water) available in addition to water for 24 hours on alternate days. After a 2-week forced abstinence, anxiety-like behavior was assessed via defensive withdrawal testing; then, IA2BC alcohol access was renewed for 2 weeks to model relapse-like behavior. RESULTS Pair-housed female rats did not increase their alcohol intake across the 8-week drinking period, unlike all other groups, and stress history did not significantly change alcohol consumption. After abstinence, anxiety-like behavior was greatest in pair-housed stress history males, whereas alcohol intake was significantly elevated only in female rats, particularly those in solitary housing. CONCLUSIONS Together, these findings suggest that paired housing differentially contributes to behavior in male and female rats, blunting alcohol intake in females, and unmasking stress history effects on anxiety-like behavior in males.
Collapse
Affiliation(s)
- Kelly M. Moench
- Department of Psychology, Indiana University – Purdue University Indianapolis, Indianapolis, IN 46202
| | - Marian L. Logrip
- Department of Psychology, Indiana University – Purdue University Indianapolis, Indianapolis, IN 46202
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
22
|
Bauer MR, Garcy DP, Boehm SL. Systemic Administration of the AMPA Receptor Antagonist, NBQX, Reduces Alcohol Drinking in Male C57BL/6J, But Not Female C57BL/6J or High-Alcohol-Preferring, Mice. Alcohol Clin Exp Res 2020; 44:2316-2325. [PMID: 32945559 DOI: 10.1111/acer.14461] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 09/04/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors are ionotropic glutamate receptors that have been investigated for their role in modulating alcohol consumption. However, little is known about the role of AMPA receptors in the control of binge-like or free-access alcohol drinking in C57BL/6J or in selectively bred high-alcohol-preferring (HAP) mice. The purpose of this experiment was to assess the role of systemic administration of the AMPA receptor antagonist, 2,3-dioxo-6-nitro-7-sulfamoyl-benzo[f]quinoxaline (NBQX), on alcohol consumption using a model of binge-like drinking, drinking in the dark (DID) and free-access 2-bottle choice (2BC) in male and female C57BL/6J and HAP mice. METHODS C57BL/6J mice were allowed free access to 20% (v/v) alcohol for 2 hours each day beginning 3 hours into the dark cycle for 4 days. On day 5, mice were intraperitoneally injected with one of 4 doses of NBQX (0, 3, 10, or 30 mg/kg; n = 10) 15 minutes before alcohol presentation and were given 4-hour alcohol access (extended DID). HAP mice were given 24-hour free access to 10% (v/v) alcohol and water for 19 days. On day 20, mice were intraperitoneally injected with one of 4 doses of NBQX (0, 3, 10, or 30 mg/kg; n = 9) 15 minutes before alcohol and water presentation. RESULTS In the first 2 hours of DID, at 30 mg/kg, male, but not female C57BL/6J or HAP, mice drank significantly less alcohol compared with controls and 30 mg/kg NBQX did not alter saccharin intake in the males. Although male HAP mice drank significantly less alcohol than female mice following 10 mg/kg NBQX, neither sex exhibited drinking that differed significantly from controls. NBQX did not reduce locomotor behavior at any dose, sex, or genotype. CONCLUSIONS These data suggest that AMPA receptors play a key role in modulating binge-like alcohol consumption without altering saccharin consumption or general locomotion and that this effect is specific to sex and genotype.
Collapse
Affiliation(s)
- Meredith R Bauer
- From the, Department of Psychology, Indiana Alcohol Research Center, Indiana University - Purdue University Indianapolis, Indianapolis, Indiana
| | - Daniel P Garcy
- From the, Department of Psychology, Indiana Alcohol Research Center, Indiana University - Purdue University Indianapolis, Indianapolis, Indiana
| | - Stephen L Boehm
- From the, Department of Psychology, Indiana Alcohol Research Center, Indiana University - Purdue University Indianapolis, Indianapolis, Indiana
| |
Collapse
|
23
|
Tan SY, Lei X, Little HC, Rodriguez S, Sarver DC, Cao X, Wong GW. CTRP12 ablation differentially affects energy expenditure, body weight, and insulin sensitivity in male and female mice. Am J Physiol Endocrinol Metab 2020; 319:E146-E162. [PMID: 32421370 PMCID: PMC7468785 DOI: 10.1152/ajpendo.00533.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Secreted hormones facilitate tissue cross talk to maintain energy balance. We previously described C1q/TNF-related protein 12 (CTRP12) as a novel metabolic hormone. Gain-of-function and partial-deficiency mouse models have highlighted important roles for this fat-derived adipokine in modulating systemic metabolism. Whether CTRP12 is essential and required for metabolic homeostasis is unknown. We show here that homozygous deletion of Ctrp12 gene results in sexually dimorphic phenotypes. Under basal conditions, complete loss of CTRP12 had little impact on male mice, whereas it decreased body weight (driven by reduced lean mass and liver weight) and improved insulin sensitivity in female mice. When challenged with a high-fat diet, Ctrp12 knockout (KO) male mice had decreased energy expenditure, increased weight gain and adiposity, elevated serum TNFα level, and reduced insulin sensitivity. In contrast, female KO mice had reduced weight gain and liver weight. The expression of lipid synthesis and catabolism genes, as well as profibrotic, endoplasmic reticulum stress, and oxidative stress genes were largely unaffected in the adipose tissue of Ctrp12 KO male mice. Despite greater adiposity and insulin resistance, Ctrp12 KO male mice fed an obesogenic diet had lower circulating triglyceride and free fatty acid levels. In contrast, lipid profiles of the leaner female KO mice were not different from those of WT controls. These data suggest that CTRP12 contributes to whole body energy metabolism in genotype-, diet-, and sex-dependent manners, underscoring complex gene-environment interactions influencing metabolic outcomes.
Collapse
Affiliation(s)
- Stefanie Y Tan
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xia Lei
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hannah C Little
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xi Cao
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
24
|
Datta U, Schoenrock SE, Bubier JA, Bogue MA, Jentsch JD, Logan RW, Tarantino LM, Chesler EJ. Prospects for finding the mechanisms of sex differences in addiction with human and model organism genetic analysis. GENES, BRAIN, AND BEHAVIOR 2020; 19:e12645. [PMID: 32012419 PMCID: PMC7060801 DOI: 10.1111/gbb.12645] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023]
Abstract
Despite substantial evidence for sex differences in addiction epidemiology, addiction-relevant behaviors and associated neurobiological phenomena, the mechanisms and implications of these differences remain unknown. Genetic analysis in model organism is a potentially powerful and effective means of discovering the mechanisms that underlie sex differences in addiction. Human genetic studies are beginning to show precise risk variants that influence the mechanisms of addiction but typically lack sufficient power or neurobiological mechanistic access, particularly for the discovery of the mechanisms that underlie sex differences. Our thesis in this review is that genetic variation in model organisms are a promising approach that can complement these investigations to show the biological mechanisms that underlie sex differences in addiction.
Collapse
Affiliation(s)
- Udita Datta
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| | - Sarah E. Schoenrock
- Center for Systems Neurogenetics of Addiction, Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Jason A. Bubier
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| | - Molly A. Bogue
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| | - James D. Jentsch
- Center for Systems Neurogenetics of Addiction, PsychologyState University of New York at BinghamtonBinghamtonNew York
| | - Ryan W. Logan
- Center for Systems Neurogenetics of Addiction, PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Lisa M. Tarantino
- Center for Systems Neurogenetics of Addiction, Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Elissa J. Chesler
- Center for Systems Neurogenetics of Addiction, The Jackson LaboratoryBar HarborMaine
| |
Collapse
|
25
|
Morrow AL, Boero G, Porcu P. A Rationale for Allopregnanolone Treatment of Alcohol Use Disorders: Basic and Clinical Studies. Alcohol Clin Exp Res 2020; 44:320-339. [PMID: 31782169 PMCID: PMC7018555 DOI: 10.1111/acer.14253] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/19/2019] [Indexed: 12/17/2022]
Abstract
For many years, research from around the world has suggested that the neuroactive steroid (3α,5α)-3-hydroxypregnan-20-one (allopregnanolone or 3α,5α-THP) may have therapeutic potential for treatment of various symptoms of alcohol use disorders (AUDs). In this critical review, we systematically address all the evidence that supports such a suggestion, delineate the etiologies of AUDs that are addressed by treatment with allopregnanolone or its precursor pregnenolone, and the rationale for treatment of various components of the disease based on basic science and clinical evidence. This review presents a theoretical framework for understanding how endogenous steroids that regulate the effects of stress, alcohol, and the innate immune system could play a key role in both the prevention and the treatment of AUDs. We further discuss cautions and limitations of allopregnanolone or pregnenolone therapy with suggestions regarding the management of risk and the potential for helping millions who suffer from AUDs.
Collapse
Affiliation(s)
- A. Leslie Morrow
- Department of Psychiatry, Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599
| | - Giorgia Boero
- Department of Psychiatry, Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599
| | - Patrizia Porcu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| |
Collapse
|
26
|
Sern KR, Fultz EK, Coelho MA, Bryant CD, Szumlinski KK. A prior history of binge-drinking increases sensitivity to the motivational valence of methamphetamine in female C57BL/6J mice. SUBSTANCE ABUSE-RESEARCH AND TREATMENT 2020; 14:1178221819897073. [PMID: 32009790 PMCID: PMC6971957 DOI: 10.1177/1178221819897073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022]
Abstract
Methamphetamine (MA) and alcohol use disorders exhibit a high degree of co-morbidity and sequential alcohol-MA mixing increases risk for co-abuse. Recently, we reported greater MA-conditioned reward in male C57BL/6J mice with a prior history of binge alcohol-drinking (14 days of 2-hour access to 5, 10, 20 and 40% alcohol). As female mice tend to binge-drink more alcohol than males and females tend to be more sensitive than males to the psychomotor-activating properties of MA, we first characterized the effects of binge-drinking upon MA-induced place-conditioning (four pairings of 0.25, 0.5, 1, 2, or 4 mg/kg IP) in females and then incorporated our prior data to analyze for sex differences in MA-conditioned reward. Prior binge-drinking history did not significantly affect locomotor hyperactivity or its sensitization in female mice. However, the dose-response function for place-conditioning was shifted to the left of water-drinking controls, indicating an increase in sensitivity to MA-conditioned reward. The examination of sex differences revealed no sex differences in alcohol intake, although females exhibited greater MA-induced locomotor stimulation than males, irrespective of their prior drinking history. No statistically significant sex difference was apparent for the potentiation of MA-conditioned reward produced by prior binge-drinking history. If relevant to humans, these data argue that both males and females with a prior binge-drinking history are similarly vulnerable to MA abuse and it remains to be determined whether or not the neural substrates underpinning this increased vulnerability reflect common or sex-specific adaptations in reward-related brain regions.
Collapse
Affiliation(s)
- Kimberly R Sern
- Department of Psychological and Brain Sciences, Developmental and Cell Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Elissa K Fultz
- Department of Psychological and Brain Sciences, Developmental and Cell Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Michal A Coelho
- Department of Psychological and Brain Sciences, Developmental and Cell Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Camron D Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, Developmental and Cell Biology, University of California Santa Barbara, Santa Barbara, CA, USA.,Department of Molecular, Developmental and Cell Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
27
|
Tchessalova D, Tronson NC. Enduring and Sex-specific Changes in Hippocampal Gene Expression after a Subchronic Immune Challenge. Neuroscience 2020; 428:76-89. [PMID: 31917350 DOI: 10.1016/j.neuroscience.2019.12.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 01/14/2023]
Abstract
Major illnesses, including heart attack and sepsis, can cause cognitive impairments, depression, and progressive memory decline that persist long after recovery from the original illness. In rodent models of sepsis or subchronic immune challenge, memory deficits also persist for weeks or months, even in the absence of ongoing neuroimmune activation. This raises the question of what mechanisms in the brain mediate such persistent changes in neural function. Here, we used RNA-sequencing as a large-scale, unbiased approach to identify changes in hippocampal gene expression long after a subchronic immune challenge previously established to cause persistent memory impairments in both males and females. We observed enduring dysregulation of gene expression three months after the end of a subchronic immune challenge. Surprisingly, there were striking sex differences in both the magnitude of changes and the specific genes and pathways altered, where males showed persistent changes in both immune- and plasticity-related genes three months after immune challenge, whereas females showed few such changes. In contrast, females showed striking differential gene expression in response to a subsequent immune challenge. Thus, immune activation has enduring and sex-specific consequences for hippocampal gene expression and the transcriptional response to subsequent stimuli. Together with findings of long-lasting memory impairments after immune challenge, these data suggest that illnesses can cause enduring vulnerability to, cognitive decline, affective disorders, and memory impairments via dysregulation of transcriptional processes in the brain.
Collapse
Affiliation(s)
- Daria Tchessalova
- Department of Psychology and Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Natalie C Tronson
- Department of Psychology and Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
28
|
Evans O, Rodríguez-Borillo O, Font L, Currie PJ, Pastor R. Alcohol Binge Drinking and Anxiety-Like Behavior in Socialized Versus Isolated C57BL/6J Mice. Alcohol Clin Exp Res 2019; 44:244-254. [PMID: 31713874 DOI: 10.1111/acer.14236] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/06/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Binge alcohol drinking has been characterized as a key feature of alcoholism. The drinking-in-the-dark (DID) preclinical model, a procedure that promotes high levels of ethanol (EtOH) intake in short periods of time, has been extensively used to investigate neuropharmacological and genetic determinants of binge-like EtOH consumption. Using DID methodology, alcohol-preferring strains of mice such as C57BL/6J (B6) mice consume enough EtOH to achieve blood concentrations (≥1.0 mg/ml) associated with behavioral intoxication (i.e., motor incoordination). DID procedures typically involve the use of socially isolated animals (single-housed prior to and during the experiment). Previous research indicates that stress associated with social isolation can induce anxiety-like behavior and promote increases in EtOH intake. The present study investigates the role of housing conditions in anxiety-like behavior and binge-like EtOH intake using a DID procedure. METHODS Male and female B6 mice were isolated or pair-housed for a period of 6 weeks prior to evaluation of anxiety-like (elevated plus maze, light and dark box, open field) and drinking (water, 10% sucrose, 10 to 30% EtOH) behavior. In order to measure intake, a variation of the standard DID procedure using a removable, transparent, and perforated plastic barrier strip (designed to temporarily divide the cage in 2) was introduced. This allowed for individual intake records (2-hour test) of isolated and socially housed animals. RESULTS Increased anxiety-like behavior and reduced sucrose consumption were found in isolated mice. The effects of housing conditions on EtOH intake were sex- and concentration-dependent. In male mice, isolation increased 20 and 30% EtOH intake. In females, however, an increased intake of EtOH (30%) was found in socialized animals. No effects of housing or sex were found at EtOH 10%. CONCLUSIONS Together with previous literature, the present study suggests that social isolation can promote anxiety-associated behavior and produce sex-dependent changes in binge-like EtOH consumption.
Collapse
Affiliation(s)
- Ophelia Evans
- Department of Psychology, Reed College, Portland, Oregon
| | | | - Laura Font
- Area de Psicobiología, Universitat Jaume I, Castellón, Spain
| | - Paul J Currie
- Department of Psychology, Reed College, Portland, Oregon
| | - Raúl Pastor
- Department of Psychology, Reed College, Portland, Oregon.,Area de Psicobiología, Universitat Jaume I, Castellón, Spain
| |
Collapse
|
29
|
Shukla SD, Restrepo R, Aroor AR, Liu X, Lim RW, Franke JD, Ford DA, Korthuis RJ. Binge Alcohol Is More Injurious to Liver in Female than in Male Rats: Histopathological, Pharmacologic, and Epigenetic Profiles. J Pharmacol Exp Ther 2019; 370:390-398. [PMID: 31262967 DOI: 10.1124/jpet.119.258871] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022] Open
Abstract
Binge alcohol consumption is a health problem, but differences between the sexes remain poorly defined. We have examined the in vivo effects of three acute, repeat binge alcohol administration on the liver in male and female rats. Sprague-Dawley rats were gavaged with alcohol (5 g/kg body weight) three times at 12-hour intervals. Blood and liver tissues were collected 4 hours after the last binge ethanol. Subsequently, several variables were analyzed. Compared with male rats, females had higher levels of blood alcohol, alanine aminotransferase, and triglycerides. Liver histology showed increased lipid vesicles that were larger in females. Protein levels of liver cytochrome P4502E1 were higher in the liver of females than in the liver of males after binge. Hepatic phospho-extracellular signal-regulated kinase 1/2 and phosph-p38 mitogen-activated protein kinase levels were lower in females compared with males after binge alcohol, but no differences were found in the phospho-C-jun N-terminal kinase levels. Peroxisome proliferator-activated receptor γ-coactivator 1α and cyclic AMP response element binding (CREB) protein levels increased more in female than in male livers; however, increases in phospho-CREB levels were lower in females. Remarkably, c-fos was reduced substantially in the livers of females, but no differences in c-myc protein were found. Binge ethanol caused elevation in acetylated (H3AcK9) and phosphoacetylated (H3AcK9PS10) histone H3 in both sexes but without any difference. Binge alcohol caused differential alterations in the levels of various species of phosphatidylethanol and a larger increase in the diacylglycerol kinase-α protein levels in the liver of female rats compared with male rats. These data demonstrate, for the first time, similarities and differences in the sex-specific responses to repeat binge alcohol leading to an increased susceptibility of female rats to have liver injury in vivo. SIGNIFICANCE STATEMENT: This study examines the molecular responses of male and female rat livers to acute binge alcohol in vivo and demonstrates significant differences in the susceptibility between sexes.
Collapse
Affiliation(s)
- Shivendra D Shukla
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Ricardo Restrepo
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Annayya R Aroor
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Xuanyou Liu
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Robert W Lim
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Jacob D Franke
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - David A Ford
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| |
Collapse
|
30
|
Mulligan MK, Lu L, Cavigelli SA, Mormède P, Terenina E, Zhao W, Williams RW, Jones BC. Impact of Genetic Variation on Stress-Related Ethanol Consumption. Alcohol Clin Exp Res 2019; 43:1391-1402. [PMID: 31034606 DOI: 10.1111/acer.14073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 04/23/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND The effect of stress on alcohol consumption in humans is highly variable, and the underlying processes are not yet understood. Attempts to model a positive relationship between stress and increased ethanol (EtOH) consumption in animals have been only modestly successful. Our hypothesis is that individual differences in stress effects on EtOH consumption are mediated by genetics. METHODS We measured alcohol consumption, using the drinking-in-the-dark (DID) paradigm in females from 2 inbred mouse strains, C57BL/6J (B6) and DBA/2J (D2), and 35 of their inbred progeny (the BXD family). A control group was maintained in normal housing and a stress group was exposed to chronic mild stress (CMS), consisting of unpredictable stressors over 7 weeks. These included predator, social, and environmental perturbations. Alcohol intake was measured over 16 weeks in both groups during baseline (preceding 5-week period), CMS (intervening 7-week period), and post-CMS (final 4-week period). RESULTS We detected a strong effect of CMS on alcohol intake. A few strains demonstrated CMS-related increased alcohol consumption; however, most showed decreased intake. We identified 1 nearly significant quantitative trait locus on chromosome 5 that contains the neuronal nitric oxide synthase gene (Nos1). The expression of Nos1 is frequently changed following alcohol exposure, and variants in this gene segregating among the BXD population may modulate alcohol intake in response to stress. CONCLUSIONS The results we present here represent the first study to combine chronic stress and alcohol consumption in a genetic reference population of mice. Differences in susceptibility to the effects of stressful environments vis-à-vis alcohol use disorders would suggest that the differences have at least some basis in genetic constitution. We have also nominated a likely candidate gene underlying the large individual differences in effects of stress on alcohol consumption.
Collapse
Affiliation(s)
- Megan K Mulligan
- The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Lu Lu
- The University of Tennessee Health Science Center, Memphis, Tennessee
| | | | - Pierre Mormède
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet-Tolosan, France
| | - Elena Terenina
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet-Tolosan, France
| | - Wenyuan Zhao
- The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Robert W Williams
- The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Byron C Jones
- The University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|