1
|
Lissek T. Enhancement of physiology via adaptive transcription. Pflugers Arch 2024:10.1007/s00424-024-03037-5. [PMID: 39482558 DOI: 10.1007/s00424-024-03037-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
The enhancement of complex physiological functions such as cognition and exercise performance in healthy individuals represents a challenging goal. Adaptive transcription programs that are naturally activated in animals to mediate cellular plasticity in response to stimulation can be leveraged to enhance physiological function above wild-type levels in young organisms and counteract complex functional decline in aging. In processes such as learning and memory and exercise-dependent muscle remodeling, a relatively small number of molecules such as certain stimulus-responsive transcription factors and immediate early genes coordinate widespread changes in cellular physiology. Adaptive transcription can be targeted by various methods including pharmaceutical compounds and gene transfer technologies. Important problems for leveraging adaptive transcription programs for physiological enhancement include a better understanding of their dynamical organization, more precise methods to influence the underlying molecular components, and the integration of adaptive transcription into multi-scale physiological enhancement concepts.
Collapse
Affiliation(s)
- Thomas Lissek
- Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| |
Collapse
|
2
|
Arjmand S, Ilaghi M, Shafie'ei M, Gobira PH, Grassi-Oliveira R, Wegener G. Exploring the potential link between ΔFosB and N-acetylcysteine in craving/relapse dynamics: can N-acetylcysteine stand out as a possible treatment candidate? Acta Neuropsychiatr 2024:1-14. [PMID: 39415655 DOI: 10.1017/neu.2024.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
From a neuroscientific point of view, one of the unique archetypes of substance use disorders is its road to relapse, in which the reward system plays a crucial role. Studies on the neurobiology of substance use disorders have highlighted the central role of a protein belonging to the Fos family of transcription factors, ΔFosB. Relying on the roles ΔFosB plays in the pathophysiology of substance use disorders, we endeavour to present some evidence demonstrating that N-acetylcysteine, a low-cost and well-tolerated over-the-counter medicine, may influence the downstream pathway of ΔFosB, thereby serving as a treatment strategy to mitigate the risk of relapse in cases of substance use.
Collapse
Affiliation(s)
- Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mehran Ilaghi
- Institute of Neuropharmacology, Kerman Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Shafie'ei
- Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Pedro H Gobira
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Rodrigo Grassi-Oliveira
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Affective Disorders, Aarhus University Hospital-Psychiatry, Aarhus, Denmark
| |
Collapse
|
3
|
Borland JM. The effects of different types of social interactions on the electrophysiology of neurons in the nucleus accumbens in rodents. Neurosci Biobehav Rev 2024; 164:105809. [PMID: 39004323 DOI: 10.1016/j.neubiorev.2024.105809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
BORLAND, J.M., The effects of different types of social interactions on the electrophysiology of neurons in the nucleus accumbens in rodents, NEUROSCI BIOBEH REV 21(1) XXX-XXX, 2024.-Sociality shapes an organisms' life. The nucleus accumbens is a critical brain region for mental health. In the following review, the effects of different types of social interactions on the physiology of neurons in the nucleus accumbens is synthesized. More specifically, the effects of sex behavior, aggression, social defeat, pair-bonding, play behavior, affiliative interactions, parental behaviors, the isolation from social interactions and maternal separation on measures of excitatory synaptic transmission, intracellular signaling and factors of transcription and translation in neurons in the nucleus accumbens in rodent models are reviewed. Similarities and differences in effects depending on the type of social interaction is then discussed. This review improves the understanding of the molecular and synaptic mechanisms of sociality.
Collapse
|
4
|
Mármol Contreras Y, Vasquez TES, Shah P, Payne K, Di Re J, Laezza F, Green TA. Bar press durations as a reliable and robust measure of frustration-related operant behavior: Sensitivity to incentive downshift and dose-response paradigms. PLoS One 2023; 18:e0296090. [PMID: 38127939 PMCID: PMC10734988 DOI: 10.1371/journal.pone.0296090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
In humans, frustrating experiences are known to trigger relapse events and individuals with higher frustration intolerance show increased risk of developing substance use disorders (SUDs). Despite this clear relationship, frustration-related behavior is seldom studied concurrently with self-administration behavior in rodent models. A major obstacle has been the lack of robust, quantitative assays of frustration-related operant behavior thus far. In previous work, we identified increased bar press (BP) durations in response to frustrating conditions in rats self-administering natural or drug rewards. Here, to propose BP durations as a measure of frustration-related behavior, we conducted an operant successive negative contrast (oSNC) study and found that increases in BP durations are observed in the absence of increased effort, providing evidence that this is a psychological phenomenon. Moreover, we assess the viability of widespread use of BP duration measurements as a behavioral tool by quantifying performance as it pertains to sensitivity, robustness, replicability, and sex differences. We conclude that increases in BP durations are a highly sensitive psychological response to frustrating conditions and that this measure is robust, replicable, and applicable to both sexes.
Collapse
Affiliation(s)
- Yorkiris Mármol Contreras
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Addiction Sciences and Therapeutics, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Mental Health Research Group, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Tileena E. S. Vasquez
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Addiction Sciences and Therapeutics, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Mental Health Research Group, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Poonam Shah
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Addiction Sciences and Therapeutics, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Mental Health Research Group, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kelsey Payne
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Addiction Sciences and Therapeutics, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Mental Health Research Group, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jessica Di Re
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Addiction Sciences and Therapeutics, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Mental Health Research Group, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Addiction Sciences and Therapeutics, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Mental Health Research Group, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Thomas A. Green
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Addiction Sciences and Therapeutics, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Mental Health Research Group, The University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
5
|
Zhou JL, de Guglielmo G, Ho AJ, Kallupi M, Pokhrel N, Li HR, Chitre AS, Munro D, Mohammadi P, Carrette LLG, George O, Palmer AA, McVicker G, Telese F. Single-nucleus genomics in outbred rats with divergent cocaine addiction-like behaviors reveals changes in amygdala GABAergic inhibition. Nat Neurosci 2023; 26:1868-1879. [PMID: 37798411 PMCID: PMC10620093 DOI: 10.1038/s41593-023-01452-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/06/2023] [Indexed: 10/07/2023]
Abstract
The amygdala processes positive and negative valence and contributes to addiction, but the cell-type-specific gene regulatory programs involved are unknown. We generated an atlas of single-nucleus gene expression and chromatin accessibility in the amygdala of outbred rats with high and low cocaine addiction-like behaviors following prolonged abstinence. Differentially expressed genes between the high and low groups were enriched for energy metabolism across cell types. Rats with high addiction index (AI) showed increased relapse-like behaviors and GABAergic transmission in the amygdala. Both phenotypes were reversed by pharmacological inhibition of the glyoxalase 1 enzyme, which metabolizes methylglyoxal-a GABAA receptor agonist produced by glycolysis. Differences in chromatin accessibility between high and low AI rats implicated pioneer transcription factors in the basic helix-loop-helix, FOX, SOX and activator protein 1 families. We observed opposite regulation of chromatin accessibility across many cell types. Most notably, excitatory neurons had greater accessibility in high AI rats and inhibitory neurons had greater accessibility in low AI rats.
Collapse
Affiliation(s)
- Jessica L Zhou
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA, USA
- Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Aaron J Ho
- Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Marsida Kallupi
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Narayan Pokhrel
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Hai-Ri Li
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Apurva S Chitre
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Daniel Munro
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Pejman Mohammadi
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - Olivier George
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Graham McVicker
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA, USA.
- Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
| | - Francesca Telese
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
6
|
Sayson LV, Ortiz DM, Lee HJ, Kim M, Custodio RJP, Yun J, Lee CH, Lee YS, Cha HJ, Cheong JH, Kim HJ. Deletion of Cryab increases the vulnerability of mice to the addiction-like effects of the cannabinoid JWH-018 via upregulation of striatal NF-κB expression. Front Pharmacol 2023; 14:1135929. [PMID: 37007015 PMCID: PMC10060981 DOI: 10.3389/fphar.2023.1135929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Synthetic cannabinoids have exhibited unpredictable abuse liabilities, especially self-administration (SA) responses in normal rodent models, despite seemingly inducing addiction-like effects in humans. Thus, an efficient pre-clinical model must be developed to determine cannabinoid abuse potential in animals and describe the mechanism that may mediate cannabinoid sensitivity. The Cryab knockout (KO) mice were recently discovered to be potentially sensitive to the addictive effects of psychoactive drugs. Herein, we examined the responses of Cryab KO mice to JWH-018 using SA, conditioned place preference, and electroencephalography. Additionally, the effects of repeated JWH-018 exposure on endocannabinoid- and dopamine-related genes in various addiction-associated brain regions were examined, along with protein expressions involving neuroinflammation and synaptic plasticity. Cryab KO mice exhibited greater cannabinoid-induced SA responses and place preference, along with divergent gamma wave alterations, compared to wild-type (WT) mice, implying their higher sensitivity to cannabinoids. Endocannabinoid- or dopamine-related mRNA expressions and accumbal dopamine concentrations after repeated JWH-018 exposure were not significantly different between the WT and Cryab KO mice. Further analyses revealed that repeated JWH-018 administration led to possibly greater neuroinflammation in Cryab KO mice, which may arise from upregulated NF-κB, accompanied by higher expressions of synaptic plasticity markers, which might have contributed to the development of cannabinoid addiction-related behavior in Cryab KO mice. These findings signify that increased neuroinflammation via NF-κB may mediate the enhanced addiction-like responses of Cryab KO mice to cannabinoids. Altogether, Cryab KO mice may be a potential model for cannabinoid abuse susceptibility.
Collapse
Affiliation(s)
- Leandro Val Sayson
- Department of Pharmacy, Uimyung Research Institute for Neuroscience, Sahmyook University, Seoul, Republic of Korea
| | - Darlene Mae Ortiz
- Department of Pharmacy, Uimyung Research Institute for Neuroscience, Sahmyook University, Seoul, Republic of Korea
| | - Hyun Jun Lee
- Department of Pharmacy, Uimyung Research Institute for Neuroscience, Sahmyook University, Seoul, Republic of Korea
| | - Mikyung Kim
- Department of Chemistry and Life Science, Sahmyook University, Seoul, Republic of Korea
| | - Raly James Perez Custodio
- Department of Ergonomics, Leibniz Research Centre for Working Environment and Human Factors—IfADo, Dortmund, Germany
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungcheongbuk-do, Republic of Korea
| | - Chae Hyeon Lee
- Medicinal Chemistry Laboratory, Department of Fundamental Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Yong Sup Lee
- Medicinal Chemistry Laboratory, Department of Fundamental Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Hye Jin Cha
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam–do, Republic of Korea
| | - Jae Hoon Cheong
- Institute for New Drug Development, School of Pharmacy, Jeonbuk National University, Jeonju, Jeollabuk-do, Republic of Korea
- *Correspondence: Jae Hoon Cheong, ; Hee Jin Kim,
| | - Hee Jin Kim
- Department of Pharmacy, Uimyung Research Institute for Neuroscience, Sahmyook University, Seoul, Republic of Korea
- *Correspondence: Jae Hoon Cheong, ; Hee Jin Kim,
| |
Collapse
|
7
|
Wang Y, Yang L, Zhou H, Zhang K, Zhao M. Identification of miRNA-mediated gene regulatory networks in L-methionine exposure counteracts cocaine-conditioned place preference in mice. Front Genet 2023; 13:1076156. [PMID: 36744178 PMCID: PMC9893020 DOI: 10.3389/fgene.2022.1076156] [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: 10/21/2022] [Accepted: 12/27/2022] [Indexed: 01/20/2023] Open
Abstract
Background and Aims: Methionine has been proven to inhibit addictive behaviors of cocaine dependence. This study aimed to identify the potential mechanisms of MET relating to its inhibitory effects on cocaine induced cellular and behavioral changes. Methods: MRNA and miRNA high-throughput sequencing of the prefrontal cortex in a mouse model of cocaine conditioned place preference (CPP) combined with L-methionine was performed. Differentially expressed miRNAs (DE-miRNAs) and differentially expressed genes (DEGs) regulated by cocaine and inhibited by L-methionine were identified. DEGs were mapped to STRING database to construct a protein-protein interaction (PPI) network. Then, the identified DEGs were subjected to the DAVID webserver for functional annotation. Finally, miRNA-mRNA regulatory network and miRNA-mRNA-TF regulatory networks were established to screen key DE-miRNAs and coregulation network in Cytoscape. Results: Sequencing data analysis showed that L-methionine reversely regulated genes and miRNAs affected by cocaine. Pathways associated with drug addiction only enriched in CS-down with MC-up genes targeted by DE-miRNAs including GABAergic synapse, Glutamatergic synapse, Circadian entrainment, Axon guidance and Calcium signaling pathway. Drug addiction associated network was formed of 22 DEGs including calcium channel (Cacna1c, Cacna1e, Cacna1g and Cacng8), ephrin receptor genes (Ephb6 and Epha8) and ryanodine receptor genes (Ryr1 and Ryr2). Calcium channel gene network were identified as a core gene network modulated by L-methionine in response to cocaine dependence. Moreover, it was predicted that Grin1 and Fosb presented in TF-miRNA-mRNA coregulation network with a high degree of interaction as hub genes and interacted calcium channels. Conclusion: These identified key genes, miRNA and coregulation network demonstrated the efficacy of L-methionine in counteracting the effects of cocaine CPP. To a certain degree, it may provide some hints to better understand the underlying mechanism on L-methionine in response to cocaine abuse.
Collapse
Affiliation(s)
- Yan Wang
- CAS Key Lab of Mental Health, Institute of Psychology, Beijing, China,Department of psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Lvyu Yang
- CAS Key Lab of Mental Health, Institute of Psychology, Beijing, China,Department of psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Hansheng Zhou
- Department of Pharmacy, Linyi People’s Hospital, Linyi, Shandong Province, China
| | - Kunlin Zhang
- CAS Key Lab of Mental Health, Institute of Psychology, Beijing, China
| | - Mei Zhao
- CAS Key Lab of Mental Health, Institute of Psychology, Beijing, China,Department of psychology, University of Chinese Academy of Sciences, Beijing, China,*Correspondence: Mei Zhao,
| |
Collapse
|
8
|
Malone SG, Shaykin JD, Stairs DJ, Bardo MT. Neurobehavioral effects of environmental enrichment and drug abuse vulnerability: An updated review. Pharmacol Biochem Behav 2022; 221:173471. [PMID: 36228739 DOI: 10.1016/j.pbb.2022.173471] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/16/2022] [Accepted: 10/05/2022] [Indexed: 12/14/2022]
Abstract
Environmental enrichment consisting of social peers and novel objects is known to alter neurobiological functioning and have an influence on the behavioral effects of drugs of abuse in preclinical rodent models. An earlier review from our laboratory (Stairs and Bardo, 2009) provided an overview of enrichment-specific changes in addiction-like behaviors and neurobiology. The current review updates the literature in this extensive field. Key findings from this updated review indicate that enrichment produces positive outcomes in drug abuse vulnerability beyond just psychostimulants. Additionally, recent studies indicate that enrichment activates key genes involved in cell proliferation and protein synthesis in nucleus accumbens and enhances growth factors in hippocampus and neurotransmitter signaling pathways in prefrontal cortex, amygdala, and hypothalamus. Remaining gaps in the literature and future directions for environmental enrichment and drug abuse research are identified.
Collapse
Affiliation(s)
- Samantha G Malone
- Department of Psychology, University of Kentucky, BBSRB, 741 S. Limestone, Lexington, KY, USA
| | - Jakob D Shaykin
- Department of Psychology, University of Kentucky, BBSRB, 741 S. Limestone, Lexington, KY, USA
| | - Dustin J Stairs
- Department of Psychological Science, Creighton University, Hixson-Lied Science Building, 2500 California Plaza, Omaha, NE, USA
| | - Michael T Bardo
- Department of Psychology, University of Kentucky, BBSRB, 741 S. Limestone, Lexington, KY, USA.
| |
Collapse
|
9
|
Han Y, Yuan M, Guo YS, Shen XY, Gao ZK, Bi X. The role of enriched environment in neural development and repair. Front Cell Neurosci 2022; 16:890666. [PMID: 35936498 PMCID: PMC9350910 DOI: 10.3389/fncel.2022.890666] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/29/2022] [Indexed: 12/01/2022] Open
Abstract
In addition to genetic information, environmental factors play an important role in the structure and function of nervous system and the occurrence and development of some nervous system diseases. Enriched environment (EE) can not only promote normal neural development through enhancing neuroplasticity but also play a nerve repair role in restoring functional activities during CNS injury by morphological and cellular and molecular adaptations in the brain. Different stages of development after birth respond to the environment to varying degrees. Therefore, we systematically review the pro-developmental and anti-stress value of EE during pregnancy, pre-weaning, and “adolescence” and analyze the difference in the effects of EE and its sub-components, especially with physical exercise. In our exploration of potential mechanisms that promote neurodevelopment, we have found that not all sub-components exert maximum value throughout the developmental phase, such as animals that do not respond to physical activity before weaning, and that EE is not superior to its sub-components in all respects. EE affects the developing and adult brain, resulting in some neuroplastic changes in the microscopic and macroscopic anatomy, finally contributing to enhanced learning and memory capacity. These positive promoting influences are particularly prominent regarding neural repair after neurobiological disorders. Taking cerebral ischemia as an example, we analyzed the molecular mediators of EE promoting repair from various dimensions. We found that EE does not always lead to positive effects on nerve repair, such as infarct size. In view of the classic issues such as standardization and relativity of EE have been thoroughly discussed, we finally focus on analyzing the essentiality of the time window of EE action and clinical translation in order to devote to the future research direction of EE and rapid and reasonable clinical application.
Collapse
Affiliation(s)
- Yu Han
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Mei Yuan
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Yi-Sha Guo
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xin-Ya Shen
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Graduate School, Shanghai University of Medicine and Health Sciences Affiliated Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhen-Kun Gao
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Graduate School, Shanghai University of Medicine and Health Sciences Affiliated Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- *Correspondence: Xia Bi
| |
Collapse
|
10
|
Sadat‐Shirazi M, Nouri Zadeh‐Tehrani S, Akbarabadi A, Mokri A, Taleb Zadeh Kasgari B, Zarrindast M. Exercise can restore behavioural and molecular changes of intergenerational morphine effects. Addict Biol 2022; 27:e13122. [PMID: 34931742 DOI: 10.1111/adb.13122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 11/28/2022]
Abstract
In our previous studies, the offspring of morphine-exposed parents (MEO) showed pharmacological tolerance to the morphine's reinforcing effect. According to the role of exercise in treatment of morphine addiction, the current study was designed to utilize exercise to improve the effect of parental morphine exposure on the morphine's reinforcing effect. Male and female rats received morphine for 10 days and were drug-free for another 10 days. Each morphine-exposed animal was allowed to mate either with a drug-naïve or a morphine-exposed rat. The offspring were divided into two groups: (1) offspring that were subjected to treadmill exercise and (2) offspring that were not subjected to exercise. The reinforcing effect of morphine was evaluated using conditioned place preference (CPP) and two-bottle choice (TBC) tests. Levels of dopamine receptors (D1DR and D2DR), μ-opioid receptor (MOR), and ΔFosB were evaluated in the nucleus accumbens. The MEO obtained lower preference scores in CPP and consumed morphine more than the control group in TBC. After 3 weeks of exercise, the reinforcing effect of morphine in the MEO was similar to the control. D1DR, D2DR, and MOR were increased in MEO compared with the controls before exercise. Levels of D1DR and MOR were decreased after exercise in the MEO; however, D1DR was increased in control. D2DR level did not change after exercise in MEO, but it increased in control group. Moreover, the level of ΔFosB was decreased among MEO while it was increased after exercise. In conclusion, exercise might modulate the reinforcing effect of morphine via alteration in levels of D1DR, MOR, and ΔFosB.
Collapse
Affiliation(s)
| | | | - Ardeshir Akbarabadi
- Iranian National Center for Addiction Studies Tehran University of Medical Sciences Tehran Iran
| | - Azarakhsh Mokri
- Iranian National Center for Addiction Studies Tehran University of Medical Sciences Tehran Iran
- Roozbeh Hospital Tehran University of Medical Sciences Tehran Iran
| | - Bahar Taleb Zadeh Kasgari
- Iranian National Center for Addiction Studies Tehran University of Medical Sciences Tehran Iran
- School of Biology, College of Science University of Tehran Tehran Iran
| | - Mohammad‐Reza Zarrindast
- Iranian National Center for Addiction Studies Tehran University of Medical Sciences Tehran Iran
- Department of Pharmacology, School of Medicine Tehran University of Medical Sciences Tehran Iran
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences Islamic Azad University Tehran Iran
- Endocrinology and Metabolism Research Institute Tehran University of Medical Science Tehran Iran
| |
Collapse
|
11
|
Cell-type- and region-specific modulation of cocaine seeking by micro-RNA-1 in striatal projection neurons. Mol Psychiatry 2022; 27:918-928. [PMID: 34785784 PMCID: PMC9054679 DOI: 10.1038/s41380-021-01328-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/16/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
The persistent and experience-dependent nature of drug addiction may result in part from epigenetic alterations, including non-coding micro-RNAs (miRNAs), which are both critical for neuronal function and modulated by cocaine in the striatum. Two major striatal cell populations, the striato-nigral and striato-pallidal projection neurons, express, respectively, the D1 (D1-SPNs) and D2 (D2-SPNs) dopamine receptor, and display distinct but complementary functions in drug-evoked responses. However, a cell-type-specific role for miRNAs action has yet to be clarified. Here, we evaluated the expression of a subset of miRNAs proposed to modulate cocaine effects in the nucleus accumbens (NAc) and dorsal striatum (DS) upon sustained cocaine exposure in mice and showed that these selected miRNAs were preferentially upregulated in the NAc. We focused on miR-1 considering the important role of some of its predicted mRNA targets, Fosb and Npas4, in the effects of cocaine. We validated these targets in vitro and in vivo. We explored the potential of miR-1 to regulate cocaine-induced behavior by overexpressing it in specific striatal cell populations. In DS D1-SPNs miR-1 overexpression downregulated Fosb and Npas4 and reduced cocaine-induced CPP reinstatement, but increased cue-induced cocaine seeking. In DS D2-SPNs miR-1 overexpression reduced the motivation to self-administer cocaine. Our results indicate a role of miR1 and its target genes, Fosb and Npas4, in these behaviors and highlight a precise cell-type- and region-specific modulatory role of miR-1, illustrating the importance of cell-specific investigations.
Collapse
|
12
|
Salinas-Velarde ID, Bernal-Morales B, Pacheco-Cabrera P, Sánchez-Aparicio P, Pascual-Mathey LI, Venebra-Muñoz A. Lower ΔFosB expression in the dopaminergic system after stevia consumption in rats housed under environmental enrichment conditions. Brain Res Bull 2021; 177:172-180. [PMID: 34624462 DOI: 10.1016/j.brainresbull.2021.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 09/12/2021] [Accepted: 10/01/2021] [Indexed: 10/20/2022]
Abstract
Environmental enrichment (EE) has been proven to reduce drug seeking and the development of addiction-related behaviors in rodent models, but the effects of EE on natural reward acquisition in the form of sweet beverages are poorly understood. Accumulating evidence shows that the intake of sugar, the main ingredient of sweet beverages, alters the dopaminergic system, leading to addiction-related physiological and molecular changes. Sugar in sweet beverages has been replaced with natural sweeteners, such as stevia extract, which has greater sweetener potential but no energy content. Our research group found that sucralose consumption increased the expression of ΔFosB in reward-related nuclei, suggesting activation of the dopaminergic system. The present study assessed the effects of EE on stevia consumption and the expression of ΔFosB in the nucleus accumbens, caudate putamen, and prefrontal cortex. Sixteen male Wistar rats, 21 days old, were randomly assigned to an EE group (n = 8) or standard environment (SE) group (n = 8) and reared for 30 days. On postnatal day 52 (PND52), the brains of four animals in each housing condition were extracted to determine basal ΔFosB levels. Stevia consumption with intermittent access and ΔFosB immunoreactivity were measured for 21 days in the remainder of the rats. Compared with SE animals, EE animals exhibited a reduction of stevia consumption and alterations of ΔFosB immunoreactivity in the reward system. These results indicate that EE reduces stevia consumption and the stevia-induced ΔFosB expression, suggesting addiction-related changes in dopaminergic nuclei, which may be interpreted as a neuroprotective effect.
Collapse
Affiliation(s)
- I Daniel Salinas-Velarde
- Laboratory of Neurobiology of Addiction and Brain Plasticity, Faculty of Sciences, Universidad Autónoma del Estado de México, State of Mexico, Mexico.
| | - Blandina Bernal-Morales
- Laboratory of Neuropharmacology, Institute for Neuroethology, Universidad Veracruzana, Xalapa, Veracruz, Mexico.
| | - Pablo Pacheco-Cabrera
- Institute for Biomedical Research, Department of Cell Biology and Physiology, Universidad Nacional Autónoma de México, Mexico City, Mexico; Institute for Neuroethology, Universidad Veracruzana, Xalapa, Veracruz, Mexico.
| | - Pedro Sánchez-Aparicio
- School of Veterinary Medicine and Zootechnics, Universidad Autónoma del Estado de México, Mexico.
| | - Luz I Pascual-Mathey
- School of Pharmaceutic Biological Chemistry, Universidad Veracruzana, Xalapa, Veracruz, Mexico.
| | - Arturo Venebra-Muñoz
- Laboratory of Neurobiology of Addiction and Brain Plasticity, Faculty of Sciences, Universidad Autónoma del Estado de México, State of Mexico, Mexico.
| |
Collapse
|
13
|
Individual Differences in Frustrative Nonreward Behavior for Sucrose in Rats Predict Motivation for Fentanyl under Progressive Ratio. eNeuro 2021; 8:ENEURO.0136-21.2021. [PMID: 34607807 PMCID: PMC8555888 DOI: 10.1523/eneuro.0136-21.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 11/21/2022] Open
Abstract
Frustrative nonreward (FN) is a construct in the Negative Valence Systems domain of the Research Domain Criteria (RDoC) from the National Institute of Mental Health. An organism’s response to frustrating situations (e.g., inability to obtain an expected reward) has broad implications for a variety of neuropsychiatric conditions, including substance use disorders. The current project developed a first of its kind rat operant behavioral model of FN based loosely on the human Point Subtraction Aggression Paradigm (PSAP). The current study shows that individual differences in FN for sucrose pellets are consistent across sessions at baseline and that the task is sensitive to reward size in male rats. More importantly, high FN behavior for sucrose predicts early “breaking” for intravenous fentanyl self-administration under a progressive ratio (PR) schedule. These results solidify frustration/ FN as an important factor for substance use disorders in addition to craving, impulsivity, and habit.
Collapse
|
14
|
Nascimento GC, de Paula BB, Ferrari DP, Iyomasa DM, Pereira YCL, Pedrazzi JF, Bortolanza M, Issy AC, Issa JPM, Leite-Panissi CRA, Iyomasa MM, Del-Bel E. Upregulation of FosB/ΔFosB in limbic circuits after tooth exodontia-induced occlusal instability in an experimental model of unpredictable chronic stress. Brain Res Bull 2021; 176:142-150. [PMID: 34500037 DOI: 10.1016/j.brainresbull.2021.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/23/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
Psychological stress and occlusal alterations are contributing etiologic factors for temporomandibular and muscular disorders in the orofacial area. The neural modulation recruited for this relationship, however, is not elucidated. The aim of this study was to investigate potential central mechanisms involved in the exodontia-induced occlusal instability associated with unpredictable chronic stress (UCS). Male adult Wistar rats were submitted to occlusal instability (unilateral molar teeth extraction) and/or to a UCS protocol and treated with diazepam or vehicle. The anxiety-like behavior was evaluated by elevated plus maze (EPM) and open field (OF) tests. Limbic structures such as the central nucleus of the amygdala (CeA), paraventricular nucleus of the hypothalamus (PVN), dorsal periaqueductal gray matter (dPAG) and nucleus accumbens core (NAc) were analyzed for expression of FosB/ΔFosB (immediate early genes) by immunohistochemistry. Exodontia and/or UCS decreased the time spent in the open arms at the EPM and the distance travelled at the OF, and increased the immobility time at the OF, suggesting anxiety-like behavior. In addition, exodontia induction resulted in an upregulation of FosB/ΔFosB in the CeA, PVN and dPAG, while UCS and exodontia + UCS upregulate FosB/ΔFosB immunoreactivity in the CeA, PVN, dPAG and NAc. Treatment with diazepam decreased the expression of FosB/ΔFosB in all analyzed structures of animals subject to UCS and exodontia + UCS, while promoted a reduction in the FosB/ΔFosB expression in the CeA, PVN and dPAG in animals subject to exodontia. Our findings showed an anxiogenic effect of exodontia and UCS, which is correlated with intranuclear neuron activation of limbic structures in a spatially dependent manner and that is prevented by the administration of diazepam.
Collapse
Affiliation(s)
- Glauce Crivelaro Nascimento
- Department of Oral and Basic Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, 14040-904, SP, Brazil
| | - Bruna Balbino de Paula
- Department of Psychology, School of Philosophy, Science and Letters of Ribeirão Preto, University of São Paulo, 14040-901, SP, Brazil
| | - Daniele P Ferrari
- Department of Neurology, School of Medicine of Ribeirão Preto, University of São Paulo, 14040-900, SP, Brazil
| | - Daniela Mizusaki Iyomasa
- Department of Psychology, School of Philosophy, Science and Letters of Ribeirão Preto, University of São Paulo, 14040-901, SP, Brazil
| | - Yamba C L Pereira
- Department of Oral and Basic Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, 14040-904, SP, Brazil
| | - João F Pedrazzi
- Department of Neurology, School of Medicine of Ribeirão Preto, University of São Paulo, 14040-900, SP, Brazil
| | - Mariza Bortolanza
- Department of Oral and Basic Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, 14040-904, SP, Brazil
| | - Ana Carolina Issy
- Department of Oral and Basic Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, 14040-904, SP, Brazil
| | - João Paulo Mardegan Issa
- Department of Oral and Basic Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, 14040-904, SP, Brazil
| | - Christie R A Leite-Panissi
- Department of Psychology, School of Philosophy, Science and Letters of Ribeirão Preto, University of São Paulo, 14040-901, SP, Brazil
| | - Mamie Mizusaki Iyomasa
- Department of Oral and Basic Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, 14040-904, SP, Brazil
| | - Elaine Del-Bel
- Department of Oral and Basic Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, 14040-904, SP, Brazil.
| |
Collapse
|
15
|
Abstract
Substance use disorders (SUDs) are chronic brain diseases characterized by transitions from recreational to compulsive drug use and aberrant drug craving that persists for months to years after abstinence is achieved. The transition to compulsive drug use implies that plasticity is occurring, altering the physiology of the brain to precipitate addicted states. Epigenetic phenomena represent a varied orchestra of transcriptional tuning mechanisms that, in response to environmental stimuli, create and maintain gene expression-mediated physiological outcomes. Therefore, epigenetic mechanisms represent a convergent regulatory framework through which the plasticity required to achieve an addicted state can arise and then persist long after drug use has ended. In the first section, we will introduce basic concepts in epigenetics, such as chromatin architecture, histones and their posttranslational modifications, DNA methylation, noncoding RNAs, and transcription factors, along with methods for their investigation. We will then examine the implications of these mechanisms in SUDs, with a particular focus on cocaine-mediated neuroepigenetic plasticity across multiple behavioral models of addiction.
Collapse
Affiliation(s)
- Andrew F Stewart
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Sasha L Fulton
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Ian Maze
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
16
|
Ortiz DM, Custodio RJP, Abiero A, Botanas CJ, Sayson LV, Kim M, Lee HJ, Kim HJ, Jeong Y, Yoon S, Lee YS, Cheong JH. The dopaminergic alterations induced by 4-F-PCP and 4-Keto-PCP may enhance their drug-induced rewarding and reinforcing effects: Implications for abuse. Addict Biol 2021; 26:e12981. [PMID: 33135332 DOI: 10.1111/adb.12981] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
Novel psychoactive substances remain the popular recreational drugs of use over the years. They continue to bypass government restrictions due to their synthesis and modifications. Recent additions to the lists are the 4-F-PCP and 4-Keto-PCP, analogs of the drug phencyclidine (PCP) known to induce adverse effects and abuse potential. However, studies on the abuse potential of 4-F-PCP and 4-Keto-PCP remain scarce. The rewarding and reinforcing effects of the drugs were assessed using conditioned place preference (CPP), self-administration, and locomotor sensitization tests. Dopamine (DA) receptor antagonists (SCH23390 and haloperidol) were administered during CPP to evaluate the involvement of the mesolimbic dopaminergic system. DA-related protein expression in the nucleus accumbens (NAcc) and ventral tegmental area (VTA) was measured. Additionally, phosphorylated cyclic-adenosine monophosphate-activated protein (AMP) response element-binding (p-CREB) protein, deltaFosB (∆FosB), and brain-derived neurotrophic factor (BDNF) protein levels in the NAcc were measured to assess the addiction neural plasticity effect of the drugs. Both 4-F-PCP and 4-Keto-PCP-induced CPP and self-administration; however, only 4-F-PCP elicited locomotor sensitization. Treatment with DA receptor antagonists (SH23390 and haloperidol) inhibited the 4-F- and 4-Keto-induced CPP. Both substances altered the levels of DA receptor D1 (DRD1), thyroxine hydroxylase (TH), DA receptor D2 (DRD2), p-CREB, ∆FosB, and BDNF. The results suggest that 4-F-PCP and 4-Keto-PCP may induce abuse potential in rodents via alterations in dopaminergic system accompanied by addiction neural plasticity.
Collapse
Affiliation(s)
- Darlene Mae Ortiz
- Uimyung Research Institute for Neuroscience, Department of Pharmacy Sahmyook University Seoul Republic of Korea
| | - Raly James Perez Custodio
- Uimyung Research Institute for Neuroscience, Department of Pharmacy Sahmyook University Seoul Republic of Korea
| | - Arvie Abiero
- Uimyung Research Institute for Neuroscience, Department of Pharmacy Sahmyook University Seoul Republic of Korea
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science University of Technology Sydney Ultimo New South Wales Australia
| | - Chrislean Jun Botanas
- Uimyung Research Institute for Neuroscience, Department of Pharmacy Sahmyook University Seoul Republic of Korea
- Department of Psychiatry University of Maryland School of Medicine Baltimore Maryland USA
| | - Leandro Val Sayson
- Uimyung Research Institute for Neuroscience, Department of Pharmacy Sahmyook University Seoul Republic of Korea
| | - Mikyung Kim
- Department of Chemistry and Life Science Sahmyook University Seoul Republic of Korea
| | - Hyun Jun Lee
- Uimyung Research Institute for Neuroscience, Department of Pharmacy Sahmyook University Seoul Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy Sahmyook University Seoul Republic of Korea
| | - Youngdo Jeong
- Medicinal Chemistry Laboratory, Department of Pharmacy and Department of Life and Nanopharmaceutical Sciences, College of Pharmacy Kyung Hee University Seoul Republic of Korea
| | - Sulmin Yoon
- School of Pharmacy Jeonbuk National University Jeonju‐si Jeollabuk‐do Republic of Korea
| | - Yong Sup Lee
- Medicinal Chemistry Laboratory, Department of Pharmacy and Department of Life and Nanopharmaceutical Sciences, College of Pharmacy Kyung Hee University Seoul Republic of Korea
| | - Jae Hoon Cheong
- Uimyung Research Institute for Neuroscience, Department of Pharmacy Sahmyook University Seoul Republic of Korea
- School of Pharmacy Jeonbuk National University Jeonju‐si Jeollabuk‐do Republic of Korea
| |
Collapse
|
17
|
Yazdanfar N, Farnam A, Sadigh-Eteghad S, Mahmoudi J, Sarkaki A. Enriched environment and social isolation differentially modulate addiction-related behaviors in male offspring of morphine-addicted dams: The possible role of μ-opioid receptors and ΔFosB in the brain reward pathway. Brain Res Bull 2021; 170:98-105. [PMID: 33592274 DOI: 10.1016/j.brainresbull.2021.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 12/20/2022]
Abstract
Prenatal opioids exposure negatively affects the neurobehavioral abilities of children born from dependence dams. Adolescent housing conditions can buffer the detrimental impacts of early life experiences or contradictory can worsen individual psychosocial functions. The present study investigated the effects of maternal morphine dependence and different rearing conditions on behaviors and protein expression in brain reward circuits of male pups. Female Wistar rats a week before conception, during pregnancy and lactation were injected twice daily with escalating doses of morphine or saline. On a postnatal day 21, male pups were weaned and subjected to three different environments for two months: standard (STD), isolated (ISO), or enriched environment (EE). The anxiety and drug-related reward were measured using elevated plus maze, open field test, and conditioned place preference. Western blotting was used to determine the protein level of ΔFosB and μ-opioid receptor proteins in the striatum and the midbrain of male offspring, respectively. Results showed that maternal morphine administration dramatically increased anxiety-like and morphine place preference behaviors in offspring. Also, ISO condition aggravated these behavioral outcomes. While, rearing in EE could attenuate anxiety and morphine conditioning in pups. At molecular levels, maternal morphine exposure and social isolation markedly increased both of ΔFosB and μ-opioid receptor proteins expression. However, rearing in the EE declined ΔFosB protein expression. Together, these findings help to elucidate long lasting impacts of early life morphine exposure and rearing environment on the behavioral and molecular profile of addicted individuals.
Collapse
Affiliation(s)
- Neda Yazdanfar
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Neuroscience, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Farnam
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Alireza Sarkaki
- The Persian Gulf Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
18
|
Crofton EJ, Nenov MN, Zhang Y, Tapia CM, Donnelly J, Koshy S, Laezza F, Green TA. Topographic transcriptomics of the nucleus accumbens shell: Identification and validation of fatty acid binding protein 5 as target for cocaine addiction. Neuropharmacology 2021; 183:108398. [PMID: 33181146 PMCID: PMC7755097 DOI: 10.1016/j.neuropharm.2020.108398] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/09/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022]
Abstract
Substance use disorders for cocaine are major public health concerns with few effective treatment options. Therefore, identification of novel pharmacotherapeutic targets is critical for future therapeutic development. Evolution has ensured that genes are expressed largely only where they are needed. Therefore, examining the gene expression landscape of the nucleus accumbens shell (NAcSh), a brain region important for reward related behaviors, may lead to the identification of novel targets for cocaine use disorder. In this study, we conducted a novel two-step topographic transcriptomic analysis using five seed transcripts with enhanced expression in the NAcSh to identify transcripts with similarly enhanced expression utilizing the correlation feature to search the more than 20,000 in situ hybridization experiments of the Allen Mouse Brain Atlas. Transcripts that correlated with at least three seed transcripts were analyzed with Ingenuity Pathway Analysis (IPA). We identified 7-fold more NAcSh-enhanced transcripts than our previous analysis using single voxels in the NAcSh as the seed. Analysis of the resulting transcripts with IPA identified many previously identified signaling pathways such as retinoic acid signaling as well as novel pathways. Manipulation of the retinoic acid pathway specifically in the NAcSh of male rats via viral vector-mediated RNA interference targeting fatty acid binding protein 5 (FABP5) decreased cocaine self-administration and modulates excitability of medium spiny neurons in the NAcSh. These results not only validate the prospective strategy of conducting a topographic transcriptomic analysis, but also further validate retinoic acid signaling as a promising pathway for pharmacotherapeutic development against cocaine use disorder.
Collapse
Affiliation(s)
- Elizabeth J Crofton
- Dept. of Pharmacology and Toxicology, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA; Neuroscience Graduate Program University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Miroslav N Nenov
- Dept. of Pharmacology and Toxicology, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Yafang Zhang
- Dept. of Pharmacology and Toxicology, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA; Pharmacology and Toxicology Graduate Program University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Cynthia M Tapia
- Dept. of Pharmacology and Toxicology, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA; Pharmacology and Toxicology Graduate Program University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Joseph Donnelly
- Dept. of Pharmacology and Toxicology, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Shyny Koshy
- Dept. of Pharmacology and Toxicology, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Fernanda Laezza
- Dept. of Pharmacology and Toxicology, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Thomas A Green
- Dept. of Pharmacology and Toxicology, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
19
|
Smail MA, Smith BL, Nawreen N, Herman JP. Differential impact of stress and environmental enrichment on corticolimbic circuits. Pharmacol Biochem Behav 2020; 197:172993. [PMID: 32659243 PMCID: PMC7484282 DOI: 10.1016/j.pbb.2020.172993] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 05/27/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022]
Abstract
Stress exposure can produce profound changes in physiology and behavior that can impair health and well-being. Of note, stress exposure is linked to anxiety disorders and depression in humans. The widespread impact of these disorders warrants investigation into treatments to mitigate the harmful effects of stress. Pharmacological treatments fail to help many with these disorders, so recent work has focused on non-pharmacological alternatives. One of the most promising of these alternatives is environmental enrichment (EE). In rodents, EE includes social, physical, and cognitive stimulation for the animal, in the form of larger cages, running wheels, and toys. EE successfully reduces the maladaptive effects of various stressors, both as treatment and prophylaxis. While we know that EE can have beneficial effects under stress conditions, the morphological and molecular mechanisms underlying these behavioral effects are still not well understood. EE is known to alter neurogenesis, dendrite development, and expression of neurotrophic growth factors, effects that vary by type of enrichment, age, and sex. To add to this complexity, EE has differential effects in different brain regions. Understanding how EE exerts its protective effects on morphological and molecular levels could hold the key to developing more targeted pharmacological treatments. In this review, we summarize the literature on the morphological and molecular consequences of EE and stress in key emotional regulatory pathways in the brain, the hippocampus, prefrontal cortex, and amygdala. The similarities and differences among these regions provide some insight into stress-EE interaction that may be exploited in future efforts toward prevention of, and intervention in, stress-related diseases.
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, United States.
| | - Brittany L Smith
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Nawshaba Nawreen
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, United States
| | - James P Herman
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States; Veterans Affairs Medical Center, Cincinnati, OH, United States; Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
20
|
Biological intersection of sex, age, and environment in the corticotropin releasing factor (CRF) system and alcohol. Neuropharmacology 2020; 170:108045. [PMID: 32217364 DOI: 10.1016/j.neuropharm.2020.108045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/13/2020] [Accepted: 03/06/2020] [Indexed: 01/21/2023]
Abstract
The neuropeptide corticotropin-releasing factor (CRF) is critical in neural circuit function and behavior, particularly in the context of stress, anxiety, and addiction. Despite a wealth of preclinical evidence for the efficacy of CRF receptor 1 antagonists in reducing behavioral pathology associated with alcohol exposure, several clinical trials have had disappointing outcomes, possibly due to an underappreciation of the role of biological variables. Although he National Institutes of Health (NIH) now mandate the inclusion of sex as a biological variable in all clinical and preclinical research, the current state of knowledge in this area is based almost entirely on evidence from male subjects. Additionally, the influence of biological variables other than sex has received even less attention in the context of neuropeptide signaling. Age (particularly adolescent development) and housing conditions have been shown to affect CRF signaling and voluntary alcohol intake, and the interaction between these biological variables is particularly relevant to the role of the CRF system in the vulnerability or resilience to the development of alcohol use disorder (AUD). Going forward, it will be important to include careful consideration of biological variables in experimental design, reporting, and interpretation. As new research uncovers conditions in which sex, age, and environment play major roles in physiological and/or pathological processes, our understanding of the complex interaction between relevant biological variables and critical signaling pathways like the CRF system in the cellular and behavioral consequences of alcohol exposure will continue to expand ultimately improving the ability of preclinical research to translate to the clinic. This article is part of the special issue on Neuropeptides.
Collapse
|
21
|
Zhang Y, Crofton EJ, Smith TES, Koshy S, Li D, Green TA. Manipulation of retinoic acid signaling in the nucleus accumbens shell alters rat emotional behavior. Behav Brain Res 2019; 376:112177. [PMID: 31449909 PMCID: PMC7359447 DOI: 10.1016/j.bbr.2019.112177] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/22/2019] [Accepted: 08/22/2019] [Indexed: 01/05/2023]
Abstract
Novel targets for depression and anxiety disorders are necessary for the development of more effective pharmacotherapeutics. Our previous study found that the retinoic acid (RA) signaling pathway is the signaling pathway most enhanced in the nucleus accumbens (NAc) shell, a region important for depression, anxiety, and addiction. Genetic manipulations of RA signaling in the NAc affecting addiction-related behavior prompted our study of the role of retinoic acid signaling in depression-related and anxiety-related behavior using in vivo RNA interference. Knockdown of the retinoic acid degradation enzyme cytochrome p450 family 26 subfamily b member 1 (Cyp26b1) in the nucleus accumbens shell increased depression-related behavior while decreasing anxiety-like behavior. Knockdown of the retinoic acid binding protein, cellular RA binding protein 2 (Crabp2), also increased depression-related behavior. Knockdown of another RA binding partner fatty acid binding protein 5 (Fabp5), did not alter these behaviors. These results further support the contention that RA signaling in the NAc shell can affect emotional behavior and that targeting some components of this pathway could be a promising avenue for developing novel treatments for depression and anxiety.
Collapse
Affiliation(s)
- Yafang Zhang
- Mental Health Research Group, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Elizabeth J Crofton
- Mental Health Research Group, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Tileena E S Smith
- Mental Health Research Group, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Shyny Koshy
- Mental Health Research Group, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Dingge Li
- Mental Health Research Group, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Thomas A Green
- Mental Health Research Group, Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
22
|
Therapeutic efficacy of environmental enrichment for substance use disorders. Pharmacol Biochem Behav 2019; 188:172829. [PMID: 31778722 DOI: 10.1016/j.pbb.2019.172829] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022]
Abstract
Addiction to drug and alcohol is regarded as a major health problem worldwide for which available treatments show limited effectiveness. The biggest challenge remains to enhance the capacities of interventions to reduce craving, prevent relapse and promote long-term recovery. New strategies to meet these challenges are being explored. Findings from preclinical work suggest that environmental enrichment (EE) holds therapeutic potential for the treatment of substance use disorders, as demonstrated in a number of animal models of drug abuse. The EE intervention introduced after drug exposure leads to attenuation of compulsive drug taking, attenuation of the rewarding (and reinforcing) effects of drugs, reductions in control of behavior by drug cues, and, very importantly, relapse prevention. Clinical work also suggests that multidimensional EE interventions (involving physical activity, social interaction, vocational training, recreational and community involvement) might produce similar therapeutic effects, if implemented continuously and rigorously. In this review we survey preclinical and clinical studies assessing the efficacy of EE as a behavioral intervention for substance use disorders and address related challenges. We also review work providing empirical evidence for EE-induced neuroplasticity within the mesocorticolimbic system that is believed to contribute to the seemingly therapeutic effects of EE on drug and alcohol-related behaviors.
Collapse
|
23
|
Sampedro-Piquero P, Ladrón de Guevara-Miranda D, Pavón FJ, Serrano A, Suárez J, Rodríguez de Fonseca F, Santín LJ, Castilla-Ortega E. Neuroplastic and cognitive impairment in substance use disorders: a therapeutic potential of cognitive stimulation. Neurosci Biobehav Rev 2019; 106:23-48. [DOI: 10.1016/j.neubiorev.2018.11.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 10/15/2018] [Accepted: 11/23/2018] [Indexed: 01/08/2023]
|
24
|
Dempsey E, Abautret-Daly Á, Docherty NG, Medina C, Harkin A. Persistent central inflammation and region specific cellular activation accompany depression- and anxiety-like behaviours during the resolution phase of experimental colitis. Brain Behav Immun 2019; 80:616-632. [PMID: 31063848 DOI: 10.1016/j.bbi.2019.05.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/18/2019] [Accepted: 05/03/2019] [Indexed: 02/08/2023] Open
Abstract
Depression and anxiety-related psychological symptoms are increasingly recognised as important co-morbidities in patients with inflammatory bowel disease (IBD). Dextran sulfate sodium (DSS) -induced colitis is an animal model of IBD in which afferent activation of the gut-brain axis can be assessed and explored as a source of behavioural change. Exposure of adult male Wistar rats to DSS (5%) in drinking water induced distal colitis. In parallel to local inflammatory responses in the gut wall, increased expression of IL-6 and iNOS was found in the cerebral cortex and an increase in ventricular volume. Immunoreactivity of immediate early gene FosB/ΔFosB activation was measured as an index of cellular activation and was increased in the nucleus accumbens and dorsal raphe nucleus in acutely colitic animals. Following resolution of the acute colitic response, sustained anhedonia in the saccharin preference test, immobility in the forced swim test, reduced burying behaviour in the marble burying test, and mild signs of anxiety in the elevated plus maze and light/dark box were observed. Central increases in iNOS expression persisted during the recovery phase and mapped to reactive microglia, particularly those found in the parenchyma surrounding circumventricular regions. Evidence of associated nitration was also found. Sustained increases in ventricular volume and reduced T2 magnetic resonance relaxometry time in cortical regions were observed during the recovery period. FosB/ΔFosB activation was evident in the dorsal raphe during recovery. Persistent central inflammation and cellular activation may underpin the emergence of symptoms of depression and anxiety in experimental colitis.
Collapse
Affiliation(s)
- Elaine Dempsey
- Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland; School of Pharmacy & Pharmaceutical Sciences, Trinity College, Dublin 2, Ireland
| | - Áine Abautret-Daly
- Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland; School of Pharmacy & Pharmaceutical Sciences, Trinity College, Dublin 2, Ireland
| | - Neil G Docherty
- Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland; Department of Physiology, School of Medicine, Trinity College, Dublin 2, Ireland
| | - Carlos Medina
- School of Pharmacy & Pharmaceutical Sciences, Trinity College, Dublin 2, Ireland; Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland; School of Pharmacy & Pharmaceutical Sciences, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
25
|
Sucrose Abstinence and Environmental Enrichment Effects on Mesocorticolimbic DARPP32 in Rats. Sci Rep 2018; 8:13174. [PMID: 30181585 PMCID: PMC6123458 DOI: 10.1038/s41598-018-29625-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/13/2018] [Indexed: 01/09/2023] Open
Abstract
Dopamine- and cAMP-regulated neuronal phosphoprotein 32 kDa (DARPP32) is a signaling molecule that could serve as a molecular switch, promoting or restraining sucrose seeking. We measured DARPP32 and pThr34 DARPP32 in the brains of male Long-Evans rats with a history of sucrose self-administration followed by 1 or 30 days of abstinence and exposure to either overnight (acute) or one month (chronic) environmental enrichment (EE). Brains were extracted following a 1 h cue reactivity test or no exposure to the test environment. Micropunches (prelimbic, infralimbic, and anterior cingulate areas of the medial prefrontal cortex, orbitofrontal cortex, dorsal striatum, nucleus accumbens, and ventral tegmental area) were then processed using Western blot. Abstinence increased, while EE decreased, sucrose seeking. DARPP32 and pThr34 DARPP32 levels were affected by testing, abstinence, and/or EE in most regions. Especially salient results were observed in the nucleus accumbens core, a region associated with relapse behaviors. Both acute and chronic EE reduced DARPP32 in the nucleus accumbens core and acute EE increased the ratio of phosphorylated to total DARPP32. Degree of DARPP32 phosphorylation negatively correlated with sucrose seeking. These findings demonstrate a potential role for DARPP32 in mediating the “anti-craving” effect of EE.
Collapse
|
26
|
Scala F, Nenov MN, Crofton EJ, Singh AK, Folorunso O, Zhang Y, Chesson BC, Wildburger NC, James TF, Alshammari MA, Alshammari TK, Elfrink H, Grassi C, Kasper JM, Smith AE, Hommel JD, Lichti CF, Rudra JS, D'Ascenzo M, Green TA, Laezza F. Environmental Enrichment and Social Isolation Mediate Neuroplasticity of Medium Spiny Neurons through the GSK3 Pathway. Cell Rep 2018; 23:555-567. [PMID: 29642012 PMCID: PMC6150488 DOI: 10.1016/j.celrep.2018.03.062] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 02/05/2018] [Accepted: 03/14/2018] [Indexed: 11/29/2022] Open
Abstract
Resilience and vulnerability to neuropsychiatric disorders are linked to molecular changes underlying excitability that are still poorly understood. Here, we identify glycogen-synthase kinase 3β (GSK3β) and voltage-gated Na+ channel Nav1.6 as regulators of neuroplasticity induced by environmentally enriched (EC) or isolated (IC) conditions-models for resilience and vulnerability. Transcriptomic studies in the nucleus accumbens from EC and IC rats predicted low levels of GSK3β and SCN8A mRNA as a protective phenotype associated with reduced excitability in medium spiny neurons (MSNs). In vivo genetic manipulations demonstrate that GSK3β and Nav1.6 are molecular determinants of MSN excitability and that silencing of GSK3β prevents maladaptive plasticity of IC MSNs. In vitro studies reveal direct interaction of GSK3β with Nav1.6 and phosphorylation at Nav1.6T1936 by GSK3β. A GSK3β-Nav1.6T1936 competing peptide reduces MSNs excitability in IC, but not EC rats. These results identify GSK3β regulation of Nav1.6 as a biosignature of MSNs maladaptive plasticity.
Collapse
Affiliation(s)
- Federico Scala
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Biophysics Graduate Program, Institute of Human Physiology, Università Cattolica, Rome, Italy
| | - Miroslav N Nenov
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Elizabeth J Crofton
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Neuroscience Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Aditya K Singh
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Oluwarotimi Folorunso
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Yafang Zhang
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Brent C Chesson
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Norelle C Wildburger
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Thomas F James
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Neuroscience Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA; Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
| | - Tahani K Alshammari
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA; Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
| | - Hannah Elfrink
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Bench Tutorials Program: Scientific Research and Design, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - James M Kasper
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Ashley E Smith
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA; Cell Biology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Jonathan D Hommel
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Cheryl F Lichti
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Jai S Rudra
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | | | - Thomas A Green
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA.
| |
Collapse
|
27
|
Kowiański P, Lietzau G, Steliga A, Czuba E, Ludkiewicz B, Waśkow M, Spodnik JH, Moryś J. Nicotine-induced CREB and DeltaFosB activity is modified by caffeine in the brain reward system of the rat. J Chem Neuroanat 2018; 88:1-12. [PMID: 29100904 DOI: 10.1016/j.jchemneu.2017.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 12/21/2022]
Abstract
Coffee and nicotine consumption are frequently combined, indicating possible intensifying effect of caffeine on smoking behavior, although neurobiological background of this phenomenon remains unknown. We aimed at determining the effect of caffeine and nicotine, applied separately or simultaneously, on activation of six structures of the brain reward system: nucleus accumbens (NAc), ventral tegmental area (VTA), amygdala (Amg), hippocampus (Hip), medial prefrontal cortex (mPfr) and dorsal striatum (CdP) in the adult male Wistar rats. Activation of two transcription factors, the phosphorylated form of cyclic AMP-response element binding protein (pCREB) and DeltaFosB (ΔFosB) was assessed by immunohistochemistry after multiple-dose five-days psychostimulants administration followed by 20min and 24h survival, respectively. Nicotine evoked the highest increase of pCREB-immunoreactivity (-ir) in NAc, while caffeine exerted the weakest effect in mPfr and CdP. Nicotine/caffeine co-administration resulted in decrease of pCREB-ir in NAc and increase in Amg, compared with the effect of each psychostimulant used separately. Nicotine was the strongest psychostimulant activating ΔFosB-ir in Amg, whereas caffeine - in Hip. Nicotine/caffeine-exerted effect upon ΔFosB-ir in Amg was weaker, whereas in mPfr stronger, than nicotine-evoked effect in these structures. In summary, pCREB and ΔFosB activation is dependent on the type of stimulus, brain structure and functional context. Activation of both transcription factors is responsible for caffeine's modifying effect upon nicotine-related behaviors and must be taken into account while quitting cigarette smoking.
Collapse
Affiliation(s)
- Przemysław Kowiański
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland; Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200 Slupsk, Poland.
| | - Grażyna Lietzau
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland; Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Aleksandra Steliga
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200 Slupsk, Poland
| | - Ewelina Czuba
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland
| | - Beata Ludkiewicz
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland
| | - Monika Waśkow
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200 Slupsk, Poland
| | - Jan H Spodnik
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland
| | - Janusz Moryś
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland
| |
Collapse
|
28
|
Ajonijebu DC, Abboussi O, Russell VA, Mabandla MV, Daniels WMU. Epigenetics: a link between addiction and social environment. Cell Mol Life Sci 2017; 74:2735-2747. [PMID: 28255755 PMCID: PMC11107568 DOI: 10.1007/s00018-017-2493-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/17/2017] [Accepted: 02/21/2017] [Indexed: 01/14/2023]
Abstract
The detrimental effects of drug abuse are apparently not limited to individuals but may also impact the vulnerability of their progenies to develop addictive behaviours. Epigenetic signatures, early life experience and environmental factors, converge to influence gene expression patterns in addiction phenotypes and consequently may serve as mediators of behavioural trait transmission between generations. The majority of studies investigating the role of epigenetics in addiction do not consider the influence of social interactions. This shortcoming in current experimental approaches necessitates developing social models that reflect the addictive behaviour in a free-living social environment. Furthermore, this review also reports on the advancement of interventions for drug addiction and takes into account the emerging roles of histone deacetylase (HDAC) inhibitors in the etiology of drug addiction and that HDAC may be a potential therapeutic target at nucleosomal level to improve treatment outcomes.
Collapse
Affiliation(s)
- Duyilemi C Ajonijebu
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Oualid Abboussi
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa.
| | - Vivienne A Russell
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Musa V Mabandla
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - William M U Daniels
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
- School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
29
|
Crofton EJ, Nenov MN, Zhang Y, Scala F, Page SA, McCue DL, Li D, Hommel JD, Laezza F, Green TA. Glycogen synthase kinase 3 beta alters anxiety-, depression-, and addiction-related behaviors and neuronal activity in the nucleus accumbens shell. Neuropharmacology 2017; 117:49-60. [PMID: 28126496 DOI: 10.1016/j.neuropharm.2017.01.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 01/15/2017] [Accepted: 01/22/2017] [Indexed: 11/24/2022]
Abstract
Psychiatric disorders such as anxiety, depression and addiction are often comorbid brain pathologies thought to share common mechanistic biology. As part of the cortico-limbic circuit, the nucleus accumbens shell (NAcSh) plays a fundamental role in integrating information in the circuit, such that modulation of NAcSh circuitry alters anxiety, depression, and addiction-related behaviors. Intracellular kinase cascades in the NAcSh have proven important mediators of behavior. To investigate glycogen-synthase kinase 3 (GSK3) beta signaling in the NAcSh in vivo we knocked down GSK3beta expression with a novel adeno-associated viral vector (AAV2) and assessed changes in anxiety- and depression-like behavior and cocaine self-administration in GSK3beta knockdown rats. GSK3beta knockdown reduced anxiety-like behavior while increasing depression-like behavior and cocaine self-administration. Correlative electrophysiological recordings in acute brain slices were used to assess the effect of AAV-shGSK3beta on spontaneous firing and intrinsic excitability of tonically active interneurons (TANs), cells required for input and output signal integration in the NAcSh and for processing reward-related behaviors. Loose-patch recordings showed that TANs transduced by AAV-shGSK3beta exhibited reduction in tonic firing and increased spike half width. When assessed by whole-cell patch clamp recordings these changes were mirrored by reduction in action potential firing and accompanied by decreased hyperpolarization-induced depolarizing sag potentials, increased action potential current threshold, and decreased maximum rise time. These results suggest that silencing of GSK3beta in the NAcSh increases depression- and addiction-related behavior, possibly by decreasing intrinsic excitability of TANs. However, this study does not rule out contributions from other neuronal sub-types.
Collapse
Affiliation(s)
- Elizabeth J Crofton
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Miroslav N Nenov
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Yafang Zhang
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Federico Scala
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA; Biophysics Graduate Program, Institute of Human Physiology, Universita Cattolica, Rome, Italy
| | - Sean A Page
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - David L McCue
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Dingge Li
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Jonathan D Hommel
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Fernanda Laezza
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Thomas A Green
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
30
|
5-HT 2C Receptor Knockdown in the Amygdala Inhibits Neuropathic-Pain-Related Plasticity and Behaviors. J Neurosci 2016; 37:1378-1393. [PMID: 28011743 DOI: 10.1523/jneurosci.2468-16.2016] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/28/2016] [Accepted: 12/20/2016] [Indexed: 12/31/2022] Open
Abstract
Neuroplasticity in the amygdala drives pain-related behaviors. The central nucleus (CeA) serves major amygdala output functions and can generate emotional-affective behaviors and modulate nocifensive responses. The CeA receives excitatory and inhibitory inputs from the basolateral nucleus (BLA) and serotonin receptor subtype 5-HT2CR in the BLA, but not CeA, has been implicated anxiogenic behaviors and anxiety disorders. Here, we tested the hypothesis that 5-HT2CR in the BLA plays a critical role in CeA plasticity and neuropathic pain behaviors in the rat spinal nerve ligation (SNL) model. Local 5-HT2CR knockdown in the BLA with stereotaxic injection of 5-HT2CR shRNA AAV vector decreased vocalizations and anxiety- and depression-like behaviors and increased sensory thresholds of SNL rats, but had no effect in sham controls. Extracellular single-unit recordings of CeA neurons in anesthetized rats showed that 5-HT2CR knockdown blocked the increase in neuronal activity (increased responsiveness, irregular spike firing, and increased burst activity) in SNL rats. At the synaptic level, 5-HT2CR knockdown blocked the increase in excitatory transmission from BLA to CeA recorded in brain slices from SNL rats using whole-cell patch-clamp conditions. Inhibitory transmission was decreased by 5-HT2CR knockdown in control and SNL conditions to a similar degree. The findings can be explained by immunohistochemical data showing increased expression of 5-HT2CR in non-GABAergic BLA cells in SNL rats. The results suggest that increased 5-HT2CR in the BLA contributes to neuropathic-pain-related amygdala plasticity by driving synaptic excitation of CeA neurons. As a rescue strategy, 5-HT2CR knockdown in the BLA inhibits neuropathic-pain-related behaviors.SIGNIFICANCE STATEMENT Neuroplasticity in the amygdala has emerged as an important pain mechanism. This study identifies a novel target and rescue strategy to control abnormally enhanced amygdala activity in an animal model of neuropathic pain. Specifically, an integrative approach of gene transfer, systems and brain slice electrophysiology, behavior, and immunohistochemistry was used to advance the novel concept that serotonin receptor subtype 5-HT2C contributes critically to the imbalance between excitatory and inhibitory drive of amygdala output neurons. Local viral vector-mediated 5-HT2CR knockdown in the amygdala normalizes the imbalance, decreases neuronal activity, and inhibits neuropathic-pain-related behaviors. The study provides valuable insight into serotonin receptor (dys)function in a limbic brain area.
Collapse
|
31
|
Lafragette A, Bardo MT, Lardeux V, Solinas M, Thiriet N. Reduction of Cocaine-Induced Locomotor Effects by Enriched Environment Is Associated with Cell-Specific Accumulation of ΔFosB in Striatal and Cortical Subregions. Int J Neuropsychopharmacol 2016; 20:237-246. [PMID: 27815415 PMCID: PMC5408985 DOI: 10.1093/ijnp/pyw097] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/02/2016] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Early exposure to enriched environments has been shown to decrease the locomotor effects induced by repeated injections of cocaine and modify basal and cocaine-induced total protein levels of the transcription factor ΔFosB in the whole striatum of mice. In this study, we aimed at characterizing whether the profile of ΔFosB accumulation induced by enriched environments and cocaine would be similar or different in terms of brain areas and cell type. METHODS We used mice expressing the eGFP protein in D1 receptor positive (D1R(+)) neurons to determine whether Δ FosB induced by enriched environment or cocaine injections (5×15 mg/kg) would occur in selective subpopulations of neurons in several subregions of the striatum and prefrontal cortex. RESULTS We found that: (1) exposure to enriched environment reduces cocaine-induced locomotor activation, confirming our previous findings; (2) exposure to enriched environment by itself increases the accumulation of Δ FosB mostly in D1R(-) cells in the shell part of the nucleus accumbens and dorsal striatum, whereas in the nucleus accumbens core, Δ FosB accumulates in both D1R(+) and D1R(-) neurons; (3) in standard environment mice, cocaine induces accumulation of Δ FosB selectively in D1R(+) cells in the nucleus accumbens, dorsal striatum, and infralimbic cortex; and (4) the effects of enriched environments and cocaine on accumulation of Δ FosB were reciprocally blocked by their combination. CONCLUSIONS Altogether, these results suggest that the enriched environment-induced reduction in behavioral effects of cocaine might result from 2 distinct effects on ΔFosB in striatal medium-sized spiny neurons belonging to the direct and indirect pathways.
Collapse
Affiliation(s)
- Audrey Lafragette
- INSERM, U-1084, Laboratory of Experimental and Clinical Neurosciences, Poitiers, France (Ms Lafragette, Ms Lardeux, Dr Solinas, and Dr Thiriet); University of Poitiers, Poitiers, France (Ms Lafragette, Ms Lardeux, Dr Solinas, and Dr Thiriet); Department of Psychology and Center for Drug Abuse Research Translation, University of Kentucky, Lexington, Kentucky (Dr Bardo).
| | - Michael T. Bardo
- INSERM, U-1084, Laboratory of Experimental and Clinical Neurosciences, Poitiers, France (Ms Lafragette, Ms Lardeux, Dr Solinas, and Dr Thiriet); University of Poitiers, Poitiers, France (Ms Lafragette, Ms Lardeux, Dr Solinas, and Dr Thiriet); Department of Psychology and Center for Drug Abuse Research Translation, University of Kentucky, Lexington, Kentucky (Dr Bardo).
| | - Virginie Lardeux
- INSERM, U-1084, Laboratory of Experimental and Clinical Neurosciences, Poitiers, France (Ms Lafragette, Ms Lardeux, Dr Solinas, and Dr Thiriet); University of Poitiers, Poitiers, France (Ms Lafragette, Ms Lardeux, Dr Solinas, and Dr Thiriet); Department of Psychology and Center for Drug Abuse Research Translation, University of Kentucky, Lexington, Kentucky (Dr Bardo).
| | - Marcello Solinas
- INSERM, U-1084, Laboratory of Experimental and Clinical Neurosciences, Poitiers, France (Ms Lafragette, Ms Lardeux, Dr Solinas, and Dr Thiriet); University of Poitiers, Poitiers, France (Ms Lafragette, Ms Lardeux, Dr Solinas, and Dr Thiriet); Department of Psychology and Center for Drug Abuse Research Translation, University of Kentucky, Lexington, Kentucky (Dr Bardo).
| | - Nathalie Thiriet
- INSERM, U-1084, Laboratory of Experimental and Clinical Neurosciences, Poitiers, France (Ms Lafragette, Ms Lardeux, Dr Solinas, and Dr Thiriet); University of Poitiers, Poitiers, France (Ms Lafragette, Ms Lardeux, Dr Solinas, and Dr Thiriet); Department of Psychology and Center for Drug Abuse Research Translation, University of Kentucky, Lexington, Kentucky (Dr Bardo).
| |
Collapse
|
32
|
Zhang Y, Kong F, Crofton EJ, Dragosljvich SN, Sinha M, Li D, Fan X, Koshy S, Hommel JD, Spratt HM, Luxon BA, Green TA. Transcriptomics of Environmental Enrichment Reveals a Role for Retinoic Acid Signaling in Addiction. Front Mol Neurosci 2016; 9:119. [PMID: 27899881 PMCID: PMC5110542 DOI: 10.3389/fnmol.2016.00119] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/25/2016] [Indexed: 11/29/2022] Open
Abstract
There exists much variability in susceptibility/resilience to addiction in humans. The environmental enrichment paradigm is a rat model of resilience to addiction-like behavior, and understanding the molecular mechanisms underlying this protective phenotype may lead to novel targets for pharmacotherapeutics to treat cocaine addiction. We investigated the differential regulation of transcript levels using RNA sequencing of the rat nucleus accumbens after environmental enrichment/isolation and cocaine/saline self-administration. Ingenuity Pathways Analysis and Gene Set Enrichment Analysis of 14,309 transcripts demonstrated that many biofunctions and pathways were differentially regulated. New functional pathways were also identified for cocaine modulation (e.g., Rho GTPase signaling) and environmental enrichment (e.g., signaling of EIF2, mTOR, ephrin). However, one novel pathway stood out above the others, the retinoic acid (RA) signaling pathway. The RA signaling pathway was identified as one likely mediator of the protective enrichment addiction phenotype, an interesting result given that nine RA signaling-related genes are expressed selectively and at high levels in the nucleus accumbens shell (NAcSh). Subsequent knockdown of Cyp26b1 (an RA degradation enzyme) in the NAcSh of rats confirmed this role by increasing cocaine self-administration as well as cocaine seeking. These results provide a comprehensive account of enrichment effects on the transcriptome and identify RA signaling as a contributing factor for cocaine addiction.
Collapse
Affiliation(s)
- Yafang Zhang
- Center for Addiction Research, The University of Texas Medical Branch, GalvestonTX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, GalvestonTX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, GalvestonTX, USA
| | - Fanping Kong
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, GalvestonTX, USA; Biomedical Informatics Program, The University of Texas Medical Branch, GalvestonTX, USA
| | - Elizabeth J Crofton
- Center for Addiction Research, The University of Texas Medical Branch, GalvestonTX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, GalvestonTX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, GalvestonTX, USA
| | - Steven N Dragosljvich
- Center for Addiction Research, The University of Texas Medical Branch, GalvestonTX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, GalvestonTX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, GalvestonTX, USA
| | - Mala Sinha
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, GalvestonTX, USA; Biomedical Informatics Program, The University of Texas Medical Branch, GalvestonTX, USA; Sealy Center for Molecular Medicine, Institute for Translational Science, The University of Texas Medical Branch, GalvestonTX, USA
| | - Dingge Li
- Center for Addiction Research, The University of Texas Medical Branch, GalvestonTX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, GalvestonTX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, GalvestonTX, USA
| | - Xiuzhen Fan
- Center for Addiction Research, The University of Texas Medical Branch, GalvestonTX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, GalvestonTX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, GalvestonTX, USA
| | - Shyny Koshy
- Center for Addiction Research, The University of Texas Medical Branch, GalvestonTX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, GalvestonTX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, GalvestonTX, USA
| | - Jonathan D Hommel
- Center for Addiction Research, The University of Texas Medical Branch, GalvestonTX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, GalvestonTX, USA
| | - Heidi M Spratt
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, GalvestonTX, USA; Biomedical Informatics Program, The University of Texas Medical Branch, GalvestonTX, USA; Sealy Center for Molecular Medicine, Institute for Translational Science, The University of Texas Medical Branch, GalvestonTX, USA; Department of Preventive Medicine and Community Health, The University of Texas Medical Branch, GalvestonTX, USA
| | - Bruce A Luxon
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, GalvestonTX, USA; Biomedical Informatics Program, The University of Texas Medical Branch, GalvestonTX, USA; Sealy Center for Molecular Medicine, Institute for Translational Science, The University of Texas Medical Branch, GalvestonTX, USA
| | - Thomas A Green
- Center for Addiction Research, The University of Texas Medical Branch, GalvestonTX, USA; Department of Pharmacology and Toxicology, The University of Texas Medical Branch, GalvestonTX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, GalvestonTX, USA
| |
Collapse
|
33
|
Zhang Y, Crofton EJ, Fan X, Li D, Kong F, Sinha M, Luxon BA, Spratt HM, Lichti CF, Green TA. Convergent transcriptomics and proteomics of environmental enrichment and cocaine identifies novel therapeutic strategies for addiction. Neuroscience 2016; 339:254-266. [PMID: 27717806 DOI: 10.1016/j.neuroscience.2016.09.051] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/21/2016] [Accepted: 09/30/2016] [Indexed: 11/25/2022]
Abstract
Transcriptomic and proteomic approaches have separately proven effective at identifying novel mechanisms affecting addiction-related behavior; however, it is difficult to prioritize the many promising leads from each approach. A convergent secondary analysis of proteomic and transcriptomic results can glean additional information to help prioritize promising leads. The current study is a secondary analysis of the convergence of recently published separate transcriptomic and proteomic analyses of nucleus accumbens (NAc) tissue from rats subjected to environmental enrichment vs. isolation and cocaine self-administration vs. saline. Multiple bioinformatics approaches (e.g. Gene Ontology (GO) analysis, Ingenuity Pathway Analysis (IPA), and Gene Set Enrichment Analysis (GSEA)) were used to interrogate these rich data sets. Although there was little correspondence between mRNA vs. protein at the individual target level, good correspondence was found at the level of gene/protein sets, particularly for the environmental enrichment manipulation. These data identify gene sets where there is a positive relationship between changes in mRNA and protein (e.g. glycolysis, ATP synthesis, translation elongation factor activity, etc.) and gene sets where there is an inverse relationship (e.g. ribosomes, Rho GTPase signaling, protein ubiquitination, etc.). Overall environmental enrichment produced better correspondence than cocaine self-administration. The individual targets contributing to mRNA and protein effects were largely not overlapping. As a whole, these results confirm that robust transcriptomic and proteomic data sets can provide similar results at the gene/protein set level even when there is little correspondence at the individual target level and little overlap in the targets contributing to the effects.
Collapse
Affiliation(s)
- Yafang Zhang
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA
| | - Elizabeth J Crofton
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA
| | - Xiuzhen Fan
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA
| | - Dingge Li
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA
| | - Fanping Kong
- Dept. of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Mala Sinha
- Sealy Center for Molecular Medicine, Institute for Translational Science, The University of Texas Medical Branch, Galveston, TX, USA
| | - Bruce A Luxon
- Dept. of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Molecular Medicine, Institute for Translational Science, The University of Texas Medical Branch, Galveston, TX, USA
| | - Heidi M Spratt
- Dept. of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Molecular Medicine, Institute for Translational Science, The University of Texas Medical Branch, Galveston, TX, USA; Dept. of Preventative Medicine and Community Health, The University of Texas Medical Branch, Galveston, TX, USA
| | - Cheryl F Lichti
- Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA
| | - Thomas A Green
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA; Dept. of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
34
|
Luo YX, Han H, Shao J, Gao Y, Yin X, Zhu WL, Han Y, Shi HS. mTOR signalling in the nucleus accumbens shell is critical for augmented effect of TFF3 on behavioural response to cocaine. Sci Rep 2016; 6:27895. [PMID: 27282818 PMCID: PMC4901260 DOI: 10.1038/srep27895] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/26/2016] [Indexed: 12/25/2022] Open
Abstract
Neuropeptides play important roles in modulating the rewarding value of abused drugs. Trefoil factor 3 (TFF3) was recently reported to modulate withdrawal syndrome of morphine, but the effects of TFF3 on the cocaine-induced behavioral changes are still elusive. In the present study, cocaine-induced hyperlocomotion and conditioned place preference (CPP) rat paradigms were provided to investigate the role of TFF3 in the reward response to cocaine. High-performance liquid chromatography (HPLC) analysis was used to analyse the dopamine concentration. The results showed that systemic TFF3 administration (0.1 mg/kg i.p.) significantly augmented cocaine- induced hyperlocomotion and CPP formation, without any effects on locomotor activity and aversive or rewarding effects per se. TFF3 significantly augmented the increment of the dopamine concentration in the NAc and the activity of the mTOR signalling pathway induced by acute cocaine exposure (10 mg/kg, i.p.) in the NAc shell, but not the core. The Intra-NAc shell infusion of rapamycin blocked TFF3-induced hyperactivity in cocaine-treatment rats. These findings indicated that TFF3 could potentiate behavioural response to cocaine, which may be associated with regulating dopamine concentration. Furthermore, the findings indicated that mTOR signalling pathway in the NAc shell is important for TFF3-induced enhancement on the cocaine-induced behavioral changes.
Collapse
Affiliation(s)
- Yi-Xiao Luo
- Department of Pharmacology, Medical School of Hunan Normal University, Changsha 410013, China
| | - Hua Han
- Department of gynecology and obstetrics, Hebei General Hospital, Shijiazhuang 050051, China
| | - Juan Shao
- Department of Senile Disease, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Yuan Gao
- Department of Biochemistry and Molecular Biology, College of basic medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Xi Yin
- Department of Functional region of Diagnosis, Hebei Medical University Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Wei-Li Zhu
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Ying Han
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Hai-Shui Shi
- Department of Biochemistry and Molecular Biology, College of basic medicine, Hebei Medical University, Shijiazhuang 050017, China.,National Institute on Drug Dependence, Peking University, Beijing 100191, China
| |
Collapse
|
35
|
Wang H, Tao X, Huang ST, Wu L, Tang HL, Song Y, Zhang G, Zhang YM. Chronic Stress Is Associated with Pain Precipitation and Elevation in DeltaFosb Expression. Front Pharmacol 2016; 7:138. [PMID: 27303299 PMCID: PMC4884751 DOI: 10.3389/fphar.2016.00138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 05/11/2016] [Indexed: 01/29/2023] Open
Abstract
A number of acute or repeated stimuli can induce expression of DeltaFosB (ΔFosB), a transcription factor derived from the fosB gene (an osteosarcoma viral oncogene) via alternative splicing. ΔFosB protein is currently viewed as a ‘molecular switch’ to repeated stimuli that gradually converts acute responses into relatively stable adaptations underlying long-term neural and behavioral plasticity. ΔFosB has received extensive attention in drug addition, depression, and stress adaptation, but changes in ΔFosB protein expression during pain is not fully understood. In this study we explored ΔFosB expression in the medial prefrontal cortex (mPFC) of rats experiencing chronic or acute stress-induced pain. Our data reveal that chronic pain induced by neonatal colorectal distension, chronic constriction injury (CCI) of the sciatic nerve, or maternal separation was associated with an increase in ΔfosB protein expression in mPFC, but acute application of acetic acid or zymosan did not alter the ΔFosB protein expression. ΔFosB expression in the rat visual cortex, a non pain-related brain region, did not change in response to (CCI) of the sciatic nerve and acetic acid treatment. In conclusion, our results indicate that ΔFosB protein expression is significantly elevated in rats that have experienced chronic pain and stress, but not acute pain. The ΔFosB protein may serve as an important transcription factor for chronic stress-induced pain. Further research is needed to improve the understanding of both the upstream signaling leading to ΔFosB protein expression as well as the regulation of ΔFosB gene expression in cortical neurons.
Collapse
Affiliation(s)
- Hang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, College of Anesthesiology, Xuzhou Medical University Xuzhou, China
| | - Xinrong Tao
- College of Medicine, Anhui University of Science and Technology Huainan, China
| | - Si-Ting Huang
- Jiangsu Province Key Laboratory of Anesthesiology, College of Anesthesiology, Xuzhou Medical University Xuzhou, China
| | - Liang Wu
- Jiangsu Province Key Laboratory of Anesthesiology, College of Anesthesiology, Xuzhou Medical University Xuzhou, China
| | - Hui-Li Tang
- Jiangsu Province Key Laboratory of Anesthesiology, College of Anesthesiology, Xuzhou Medical University Xuzhou, China
| | - Ying Song
- Jiangsu Province Key Laboratory of Anesthesiology, College of Anesthesiology, Xuzhou Medical University Xuzhou, China
| | - Gongliang Zhang
- School of Basic Medical Sciences, Anhui Medical University Hefei, China
| | - Yong-Mei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, College of Anesthesiology, Xuzhou Medical University Xuzhou, China
| |
Collapse
|
36
|
van der Veen R, Kentrop J, van der Tas L, Loi M, van IJzendoorn MH, Bakermans-Kranenburg MJ, Joëls M. Complex Living Conditions Impair Behavioral Inhibition but Improve Attention in Rats. Front Behav Neurosci 2015; 9:357. [PMID: 26733839 PMCID: PMC4689791 DOI: 10.3389/fnbeh.2015.00357] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/08/2015] [Indexed: 01/01/2023] Open
Abstract
Rapid adaptation to changes, while maintaining a certain level of behavioral inhibition is an important feature in every day functioning. How environmental context and challenges in life can impact on the development of this quality is still unknown. In the present study, we examined the effect of a complex rearing environment during adolescence on attention and behavioral inhibition in adult male rats. We also tested whether these effects were affected by an adverse early life challenge, maternal deprivation (MD). We found that animals that were raised in large, two floor MarlauTM cages, together with 10 conspecifics, showed improved attention, but impaired behavioral inhibition in the 5-choice serial reaction time task. The early life challenge of 24 h MD on postnatal day 3 led to a decline in bodyweight during adolescence, but did not by itself influence responses in the 5-choice task in adulthood, nor did it moderate the effects of complex housing. Our data suggest that a complex rearing environment leads to a faster adaptation to changes in the environment, but at the cost of lower behavioral inhibition.
Collapse
Affiliation(s)
- Rixt van der Veen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands; Centre for Child and Family Studies, Leiden UniversityLeiden, Netherlands
| | - Jiska Kentrop
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | - Liza van der Tas
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | - Manila Loi
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | | | | | - Marian Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| |
Collapse
|
37
|
Love T, Laier C, Brand M, Hatch L, Hajela R. Neuroscience of Internet Pornography Addiction: A Review and Update. Behav Sci (Basel) 2015; 5:388-433. [PMID: 26393658 PMCID: PMC4600144 DOI: 10.3390/bs5030388] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/29/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023] Open
Abstract
Many recognize that several behaviors potentially affecting the reward circuitry in human brains lead to a loss of control and other symptoms of addiction in at least some individuals. Regarding Internet addiction, neuroscientific research supports the assumption that underlying neural processes are similar to substance addiction. The American Psychiatric Association (APA) has recognized one such Internet related behavior, Internet gaming, as a potential addictive disorder warranting further study, in the 2013 revision of their Diagnostic and Statistical Manual. Other Internet related behaviors, e.g., Internet pornography use, were not covered. Within this review, we give a summary of the concepts proposed underlying addiction and give an overview about neuroscientific studies on Internet addiction and Internet gaming disorder. Moreover, we reviewed available neuroscientific literature on Internet pornography addiction and connect the results to the addiction model. The review leads to the conclusion that Internet pornography addiction fits into the addiction framework and shares similar basic mechanisms with substance addiction. Together with studies on Internet addiction and Internet Gaming Disorder we see strong evidence for considering addictive Internet behaviors as behavioral addiction. Future research needs to address whether or not there are specific differences between substance and behavioral addiction.
Collapse
Affiliation(s)
- Todd Love
- Society for the Advancement of Sexual Health, Ardmore, PA 19003, USA.
| | - Christian Laier
- Department of General Psychology: Cognition, University of Duisburg-Essen, Duisburg 47057, Germany.
| | - Matthias Brand
- Department of General Psychology: Cognition, University of Duisburg-Essen, Duisburg 47057, Germany.
- Erwin L. Hahn Institute for Magnetic Resonance Imaging, Essen 45141, Germany.
| | - Linda Hatch
- Private Practice, Santa Barbara, CA 93103, USA.
| | - Raju Hajela
- Health Upwardly Mobile Inc., Calgary, AB T2S 0J2, Canada.
- Diagnostic and Descriptive Terminology Action Group (DDTAG), American Society of Addiction Medicine (ASAM), Chevy Chase, MD 93101, USA.
| |
Collapse
|
38
|
Ménard C, Hodes GE, Russo SJ. Pathogenesis of depression: Insights from human and rodent studies. Neuroscience 2015; 321:138-162. [PMID: 26037806 DOI: 10.1016/j.neuroscience.2015.05.053] [Citation(s) in RCA: 370] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/14/2015] [Accepted: 05/21/2015] [Indexed: 12/30/2022]
Abstract
Major depressive disorder (MDD) will affect one out of every five people in their lifetime and is the leading cause of disability worldwide. Nevertheless, mechanisms associated with the pathogenesis of MDD have yet to be completely understood and current treatments remain ineffective in a large subset of patients. In this review, we summarize the most recent discoveries and insights for which parallel findings have been obtained in human depressed subjects and rodent models of mood disorders in order to examine the potential etiology of depression. These mechanisms range from synaptic plasticity mechanisms to epigenetics and the immune system where there is strong evidence to support a functional role in the development of specific depression symptomology. Ultimately we conclude by discussing how novel therapeutic strategies targeting central and peripheral processes might ultimately aid in the development of effective new treatments for MDD and related stress disorders.
Collapse
Affiliation(s)
- C Ménard
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - G E Hodes
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - S J Russo
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
39
|
Nestler EJ. ∆FosB: a transcriptional regulator of stress and antidepressant responses. Eur J Pharmacol 2015; 753:66-72. [PMID: 25446562 PMCID: PMC4380559 DOI: 10.1016/j.ejphar.2014.10.034] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 09/11/2014] [Accepted: 10/09/2014] [Indexed: 02/08/2023]
Abstract
ΔFosB is a member of the Fos family of transcription factors. While other family members are induced rapidly but transiently in response to a host of acute stimuli, ΔFosB is unique in that it accumulates in response to repeated stimulation due to its unusual protein stability. Such prolonged induction of ΔFosB, within nucleus accumbens (NAc), a key brain reward region, has been most studied in animal models of drug addiction, with considerable evidence indicating that ΔFosB promotes reward and motivation and serves as a mechanism of drug sensitization and increased drug self-administration. In more recent years, prolonged induction of ∆FosB has also been observed within NAc in response to chronic administration of certain forms of stress. Increasing evidence indicates that this induction represents a positive, homeostatic adaptation to chronic stress, since overexpression of ∆FosB in this brain region promotes resilience to stress, whereas blockade of its activity promotes stress susceptibility. Chronic administration of several antidepressant medications also induces ∆FosB in the NAc, and this induction is required for the therapeutic-like actions of these drugs in mouse models. Validation of these rodent findings is the demonstration that depressed humans, examined at autopsy, display reduced levels of ∆FosB within the NAc. As a transcription factor, ΔFosB produces this behavioral phenotype by regulating the expression of specific target genes, which are under current investigation. These studies of ΔFosB are providing new insight into the molecular basis of depression and antidepressant action, which is defining a host of new targets for possible therapeutic development.
Collapse
Affiliation(s)
- Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
40
|
Crofton EJ, Zhang Y, Green TA. Inoculation stress hypothesis of environmental enrichment. Neurosci Biobehav Rev 2015; 49:19-31. [PMID: 25449533 PMCID: PMC4305384 DOI: 10.1016/j.neubiorev.2014.11.017] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/28/2014] [Accepted: 11/21/2014] [Indexed: 01/28/2023]
Abstract
One hallmark of psychiatric conditions is the vast continuum of individual differences in susceptibility vs. resilience resulting from the interaction of genetic and environmental factors. The environmental enrichment paradigm is an animal model that is useful for studying a range of psychiatric conditions, including protective phenotypes in addiction and depression models. The major question is how environmental enrichment, a non-drug and non-surgical manipulation, can produce such robust individual differences in such a wide range of behaviors. This paper draws from a variety of published sources to outline a coherent hypothesis of inoculation stress as a factor producing the protective enrichment phenotypes. The basic tenet suggests that chronic mild stress from living in a complex environment and interacting non-aggressively with conspecifics can inoculate enriched rats against subsequent stressors and/or drugs of abuse. This paper reviews the enrichment phenotypes, mulls the fundamental nature of environmental enrichment vs. isolation, discusses the most appropriate control for environmental enrichment, and challenges the idea that cortisol/corticosterone equals stress. The intent of the inoculation stress hypothesis of environmental enrichment is to provide a scaffold with which to build testable hypotheses for the elucidation of the molecular mechanisms underlying these protective phenotypes and thus provide new therapeutic targets to treat psychiatric/neurological conditions.
Collapse
Affiliation(s)
- Elizabeth J Crofton
- Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States
| | - Yafang Zhang
- Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States
| | - Thomas A Green
- Center for Addiction Research, Mitchell Center for Neurodegenerative Diseases, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States.
| |
Collapse
|