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
|
Bergkamp DJ, Neumaier JF. How omics is revealing new roles for glia in addiction. Glia 2024. [PMID: 38894643 DOI: 10.1002/glia.24584] [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: 01/23/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024]
Abstract
Experiments to study the biology of addiction have historically focused on the mechanisms through which drugs of abuse drive changes in the functioning of neurons and neural circuits. Glia have often been ignored in these studies, however, and this has left many questions in the field unanswered, particularly, surrounding how glia contribute to changes in synaptic plasticity, regulation of neuroinflammation, and functioning of neural ensembles given massive changes in signaling across the CNS. Omics methods (transcriptomics, translatomics, epigenomics, proteomics, metabolomics, and others) have expanded researchers' abilities to generate hypotheses and carry out mechanistic studies of glial cells during acquisition of drug taking, intoxication, withdrawal, and relapse to drug seeking. Here, we present a survey of how omics technological advances are revising our understanding of astrocytes, microglia, oligodendrocytes, and ependymal cells in addiction biology.
Collapse
Affiliation(s)
- David J Bergkamp
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
- VISN 20 Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
| | - John F Neumaier
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
- VISN 20 Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, Washington, USA
| |
Collapse
|
3
|
Hauser SR, Waeiss RA, Deehan GA, Engleman EA, Bell RL, Rodd ZA. Adolescent alcohol and nicotine exposure alters the adult response to alcohol use. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2023; 3:11880. [PMID: 38389816 PMCID: PMC10880795 DOI: 10.3389/adar.2023.11880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/31/2023] [Indexed: 02/24/2024]
Abstract
Adolescence through young adulthood is a unique period of neuronal development and maturation. Numerous agents can alter this process, resulting in long-term neurological and biological consequences. In the clinical literature, it is frequently reported that adolescent alcohol consumption increases the propensity to develop addictions, including alcohol use disorder (AUD), during adulthood. A general limitation of both clinical and human pre-clinical adolescent alcohol research is the high rate of co-using/abusing more than one drug during adolescence, such as co-using/abusing alcohol with nicotine. A primary goal of basic research is elucidating neuroadaptations produced by adolescent alcohol exposure/consumption that promote alcohol and other drug self-administration in adulthood. The long-term goal is to develop pharmacotherapeutics for the prevention or amelioration of these neuroadaptations. This review will focus on studies that have examined the effects of adolescent alcohol and nicotine exposure on adult alcohol consumption, the hypersensitivity of the mesolimbic dopaminergic system, and enhanced responses not only to alcohol but also to nicotine during adulthood. Again, the long-term goal is to identify potential cholinergic agents to prevent or ameliorate the consequences of, peri-adolescent alcohol abuse.
Collapse
Affiliation(s)
- Sheketha R Hauser
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Robert A Waeiss
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Gerald A Deehan
- Department of Psychology, East Tennessee State University, Johnson City, TN, United States
| | - Eric A Engleman
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Richard L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Zachary A Rodd
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
4
|
Chen Z, Chen W, Li Y, Moos M, Xiao D, Wang C. Single-nucleus chromatin accessibility and RNA sequencing reveal impaired brain development in prenatally e-cigarette exposed neonatal rats. iScience 2022; 25:104686. [PMID: 35874099 PMCID: PMC9304611 DOI: 10.1016/j.isci.2022.104686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/13/2022] [Accepted: 06/24/2022] [Indexed: 11/03/2022] Open
Abstract
Although emerging evidence reveals that vaping alters the function of the central nervous system, the effects of maternal vaping on offspring brain development remain elusive. Using a well-established in utero exposure model, we performed single-nucleus ATAC-seq (snATAC-seq) and RNA sequencing (snRNA-seq) on prenatally e-cigarette-exposed rat brains. We found that maternal vaping distorted neuronal lineage differentiation in the neonatal brain by promoting excitatory neurons and inhibiting lateral ganglionic eminence-derived inhibitory neuronal differentiation. Moreover, maternal vaping disrupted calcium homeostasis, induced microglia cell death, and elevated susceptibility to cerebral ischemic injury in the developing brain of offspring. Our results suggest that the aberrant calcium signaling, diminished microglial population, and impaired microglia-neuron interaction may all contribute to the underlying mechanisms by which prenatal e-cigarette exposure impairs neonatal rat brain development. Our findings raise the concern that maternal vaping may cause adverse long-term brain damage to the offspring.
Collapse
Affiliation(s)
- Zhong Chen
- Center for Genomics, School of Medicine, Loma Linda University, 11021 Campus St., Loma Linda, CA 92350, USA
| | - Wanqiu Chen
- Center for Genomics, School of Medicine, Loma Linda University, 11021 Campus St., Loma Linda, CA 92350, USA
| | - Yong Li
- Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Malcolm Moos
- Center for Biologics Evaluation and Research & Division of Cellular and Gene Therapies, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Daliao Xiao
- Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Charles Wang
- Center for Genomics, School of Medicine, Loma Linda University, 11021 Campus St., Loma Linda, CA 92350, USA
- Division of Microbiology & Molecular Genetics, Department of Basic Science, School of Medicine, Loma Linda University, 11021 Campus St., Loma Linda, CA 92350, USA
| |
Collapse
|
5
|
Chen G, Ghazal M, Rahman S, Lutfy K. The impact of adolescent nicotine exposure on alcohol use during adulthood: The role of neuropeptides. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:53-93. [PMID: 34801174 DOI: 10.1016/bs.irn.2021.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Nicotine and alcohol abuse and co-dependence represent major public health crises. Indeed, previous research has shown that the prevalence of alcoholism is higher in smokers than in non-smokers. Adolescence is a susceptible period of life for the initiation of nicotine and alcohol use and the development of nicotine-alcohol codependence. However, there is a limited number of pharmacotherapeutic agents to treat addiction to nicotine or alcohol alone. Notably, there is no effective medication to treat this comorbid disorder. This chapter aims to review the early nicotine use and its impact on subsequent alcohol abuse during adolescence and adulthood as well as the role of neuropeptides in this comorbid disorder. The preclinical and clinical findings discussed in this chapter will advance our understanding of this comorbid disorder's neurobiology and lay a foundation for developing novel pharmacotherapies to treat nicotine and alcohol codependence.
Collapse
Affiliation(s)
- G Chen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States; Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - M Ghazal
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - S Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, United States
| | - K Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States.
| |
Collapse
|
6
|
DeBaker MC, Robinson JM, Moen JK, Wickman K, Lee AM. Differential patterns of alcohol and nicotine intake: Combined alcohol and nicotine binge consumption behaviors in mice. Alcohol 2020; 85:57-64. [PMID: 31557515 DOI: 10.1016/j.alcohol.2019.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/30/2019] [Accepted: 09/16/2019] [Indexed: 02/08/2023]
Abstract
Late adolescence and young adulthood, corresponding to the high school and college years, are vulnerable periods for increased alcohol and nicotine use. The dramatic increase in the prevalence of electronic cigarette use is particularly concerning in these age groups. Late adolescents and young adults are more likely to engage in cycles of binge drug consumption, and alcohol and nicotine are frequently used together. However, there are few data examining the combination of alcohol and nicotine in binge models in animal models. In this study, our objectives were to determine how voluntary nicotine consumption beginning in late adolescence influenced subsequent binge alcohol consumption in young adulthood, how a combination of alcohol and nicotine binge consumption differed from alcohol-only binge consumption, and whether nicotine would be consumed when presented in a binge procedure. Male C57BL/6J mice voluntarily consumed unsweetened alcohol and nicotine in continuous-access bottle-choice procedures in combination with cycles of drinking-in-the-dark. Our results show that experience with voluntary nicotine consumption in late adolescence did not affect subsequent binge alcohol consumption in early adulthood. However, mice that consumed nicotine in adolescence showed an initial decrease in alcohol preference, and consequently increase in nicotine preference, on the first session of combined ethanol and nicotine binge consumption in adulthood compared with mice that drank only water during late adolescence. Lastly, we found that mice readily consumed unsweetened nicotine when presented in a binge procedure, and the level of consumption exceeded the nicotine consumption observed in the combination alcohol and nicotine binge. Our data show that expansion of the patterns of alcohol and nicotine co-consumption in a mouse models is possible, which will enable us to dissect relevant molecular targets underlying these consumption patterns and better inform drug development efforts.
Collapse
|
7
|
Effects of the synthetic psychedelic 2,5-dimethoxy-4-iodoamphetamine (DOI) on ethanol consumption and place conditioning in male mice. Psychopharmacology (Berl) 2019; 236:3567-3578. [PMID: 31309240 PMCID: PMC6895420 DOI: 10.1007/s00213-019-05328-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 07/08/2019] [Indexed: 12/27/2022]
Abstract
RATIONALE Approximately 20 million adults in the USA have an alcohol use disorder (AUD). There are clinical and preclinical data suggesting that psychedelics may have benefits for AUD. OBJECTIVE To investigate the effects of the synthetic psychedelic 2,5-dimethoxy-4-iodoamphetamine (DOI) on the behavioral effects of ethanol. METHODS The effects of DOI were examined using ethanol-induced place conditioning (1.8 g/kg ethanol) and 2-bottle choice ethanol drinking (20% v/v), using a dose of DOI (3 mg/kg) that produced the maximal response in the serotonin 2A (5-HT2A) receptor-dependent head-twitch assay. Interactions between DOI and ethanol (3 g/kg) were examined using the ethanol-induced loss of righting reflex procedure and blood-ethanol analysis. To examine additional mechanisms by which psychedelics may interact with ethanol, we determined whether DOI reverses ethanol-induced nitric oxide release in macrophages, a marker of inflammation. RESULTS DOI significantly attenuated ethanol-induced place conditioning and ethanol drinking. DOI-induced suppression of alcohol drinking depended upon 5-HT2A receptors, was selective for alcohol over water, and was selective for high alcohol-preferring subjects. DOI had no apparent pharmacokinetic interactions with ethanol, and DOI reduced ethanol-induced nitric oxide release. CONCLUSIONS Our findings demonstrate that DOI blocks ethanol place conditioning and selectively reduces voluntary ethanol consumption. This may be related to modulation of the effects of ethanol in the reward circuitry of the brain, ethanol-induced neuroinflammation, or a combination of both. Additional studies to elucidate the mechanisms through which psychedelics attenuate the effects of ethanol would inform the pathophysiology of AUD and potentially provide new treatment options.
Collapse
|
8
|
Caruso MJ, Seemiller LR, Fetherston TB, Miller CN, Reiss DE, Cavigelli SA, Kamens HM. Adolescent social stress increases anxiety-like behavior and ethanol consumption in adult male and female C57BL/6J mice. Sci Rep 2018; 8:10040. [PMID: 29968802 PMCID: PMC6030218 DOI: 10.1038/s41598-018-28381-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/21/2018] [Indexed: 02/07/2023] Open
Abstract
Exposure to social stress is an important risk factor for comorbid affective disorders and problem alcohol use. To better understand mechanisms involved in social stress-induced affective disorder and alcohol use co-morbidity, we studied the effects of adolescent social stress on anxiety- and depression-like behaviors and binge-like ethanol consumption. Male and female C57BL/6J mice were exposed to chronic variable social stress (CVSS) or control conditions throughout adolescence (postnatal days, PND, 25-59) and then tested for anxiety-like behavior in the elevated plus maze and a novel open field environment, or depression-like behavior using the forced swim test on PND 64-66. Mice were then tested for binge-like ethanol consumption using the Drinking-in-the-Dark model. Male and female mice exposed to adolescent CVSS had increased adult anxiety-like behavior and increased locomotor adaptation to a novel environment. Further, CVSS mice consumed significantly more ethanol, but not saccharin, than controls. Despite group differences in both anxiety-like behavior and ethanol consumption, there was no relationship between these outcomes within individual mice. These data suggest that exposure to adolescent social stress is an important risk factor for later alcohol use and affective behaviors, but that social stress does not necessarily dictate co-morbidity of these outcomes.
Collapse
Affiliation(s)
- M J Caruso
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, 16802, USA.,Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - L R Seemiller
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, 16802, USA.,The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - T B Fetherston
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, 16802, USA
| | - C N Miller
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, 16802, USA
| | - D E Reiss
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, 16802, USA
| | - S A Cavigelli
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, 16802, USA.,The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - H M Kamens
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|