101
|
Merkel SF, Andrews AM, Lutton EM, Razmpour R, Cannella LA, Ramirez SH. Dexamethasone Attenuates the Enhanced Rewarding Effects of Cocaine Following Experimental Traumatic Brain Injury. Cell Transplant 2018; 26:1178-1192. [PMID: 28933216 PMCID: PMC5447499 DOI: 10.1177/0963689717714341] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Clinical studies have identified traumatic brain injury (TBI) as a risk factor for the development of cocaine dependence. This claim is supported by our recent preclinical studies showing enhancement of the rewarding effects of cocaine in mice sustaining moderate controlled cortical impact (CCI) injury during adolescence. Here we test the efficacy of dexamethasone, an anti-inflammatory corticosteroid, to attenuate augmentation of the behavioral response to cocaine observed in CCI-TBI animals using the conditioned place preference (CPP) assay. These studies were performed in order to determine whether proinflammatory activity in the nucleus accumbens (NAc), a key brain nucleus in the reward pathway, mediates enhanced cocaine-induced CPP in adolescent animals sustaining moderate CCI-TBI. Our data reveal robust glial activation in the NAc following CCI-TBI and a significant increase in the cocaine-induced CPP of untreated CCI-TBI mice. Furthermore, our results show that dexamethasone treatment following CCI-TBI can attenuate the cocaine place preference of injured animals without producing aversion in the CPP assay. Our studies also found that dexamethasone treatment significantly reduced the expression of select immune response genes including Monocyte chemoattractant protein-1 (MCP-1/CCL2) and intercellular adhesion molecule-1 ( ICAM-1), returning their expression to control levels, which prompted an investigation of peripheral blood monocytes in dexamethasone-treated animals. Experimental findings showed that no craniectomy/dexamethasone mice had a significant increase, while CCI-TBI/dexamethasone animals had a significant decrease in the percentage of circulating nonclassical patrolling monocytes. These results suggest that a portion of these monocytes may migrate to the brain in response to CCI-TBI, potentially sparing the development of chronic neuroinflammation in regions associated with the reward circuitry such as the NAc. Overall, our findings indicate that anti-inflammatory agents, such as dexamethasone, may be effective in normalizing the rewarding effects of cocaine following CCI-TBI.
Collapse
Affiliation(s)
- Steven F Merkel
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,2 Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Allison M Andrews
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,2 Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Evan M Lutton
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Roshanak Razmpour
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Lee Anne Cannella
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,2 Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Servio H Ramirez
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,2 Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,3 Shriners Hospitals Pediatric Research Center, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
102
|
Zhu R, Bu Q, Fu D, Shao X, Jiang L, Guo W, Chen B, Liu B, Hu Z, Tian J, Zhao Y, Cen X. Toll-like receptor 3 modulates the behavioral effects of cocaine in mice. J Neuroinflammation 2018; 15:93. [PMID: 29571298 PMCID: PMC5865345 DOI: 10.1186/s12974-018-1130-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 03/13/2018] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The nucleus accumbens in the midbrain dopamine limbic system plays a key role in cocaine addiction. Toll-like receptors (TLRs) are important pattern-recognition receptors (PPRs) in the innate immune system that are also involved in drug dependence; however, the detailed mechanism is largely unknown. METHODS The present study was designed to investigate the potential role of TLR3 in cocaine addiction. Cocaine-induced conditioned place preference (CPP), locomotor activity, and self-administration were used to determine the effects of TLR3 in the rewarding properties of cocaine. Lentivirus-mediated re-expression of Tlr3 (LV-TLR3) was applied to determine if restoration of TLR3 expression in the NAc is sufficient to restore the cocaine effect in TLR3-/- mice. The protein levels of phospho-NF-κB p65, IKKβ, and p-IκBα both in the cytoplasm and nucleus of cocaine-induced CPP mice were detected by Western blot. RESULTS We showed that both TLR3 deficiency and intra-NAc injection of TLR3 inhibitors significantly attenuated cocaine-induced CPP, locomotor activity, and self-administration in mice. Importantly, the TLR3-/- mice that received intra-NAc injection of LV-TLR3 displayed significant increases in cocaine-induced CPP and locomotor activity. Finally, we found that TLR3 inhibitor reverted cocaine-induced upregulation of phospho-NF-κB p65, IKKβ, and p-IκBα. CONCLUSIONS Taken together, our results describe that TLR3 modulates cocaine-induced behaviors and provide further evidence supporting a role for central pro-inflammatory immune signaling in drug reward. We propose that TLR3 blockade could be a novel approach to treat cocaine addiction.
Collapse
Affiliation(s)
- Ruiming Zhu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Qian Bu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China.,Healthy Food Evaluation Research Center, Department of Food Science and Technology, College of Light Industry, Textile and Food Engineering, Sichuan University, Chengdu, 610065, China
| | - Dengqi Fu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Xue Shao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Linhong Jiang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Wei Guo
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China.,School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Bo Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Bin Liu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Zhengtao Hu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Jingwei Tian
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Yinglan Zhao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China.
| |
Collapse
|
103
|
Karelina K, Nicholson S, Weil ZM. Minocycline blocks traumatic brain injury-induced alcohol consumption and nucleus accumbens inflammation in adolescent male mice. Brain Behav Immun 2018; 69:532-539. [PMID: 29395778 PMCID: PMC6698899 DOI: 10.1016/j.bbi.2018.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 12/26/2022] Open
Abstract
Alcohol use is a well characterized risk factor for traumatic brain injury (TBI); however, emerging clinical and experimental research suggests that TBI may also be an independent risk factor for the development of alcohol use disorders. In particular, TBIs incurred early in life predict the development of problem alcohol use and increase vulnerability to neuroinflammation as a consequence of alcohol use. Critically, the neuroinflammatory response to alcohol, mediated in large part by microglia, may also function as a driver of further alcohol use. Here, we tested the hypothesis that TBI increases alcohol consumption through microglia-mediated neuroinflammation. Mice were injured as juveniles and alcohol consumption and preference were assessed in a free-choice voluntary drinking paradigm in adolescence. TBI increased alcohol consumption; however, treatment with minocycline, an inhibitor of microglial activation, reduced alcohol intake in TBI mice to sham levels. Moreover, a single injection of ethanol (2 g/kg) significantly increased microglial activation in the nucleus accumbens and microglial expression of the proinflammatory cytokine IL-1β in TBI, but not sham or minocycline-treated, mice. Our data implicate TBI-induced microglial activation as a possible mechanism for the development of alcohol use disorders.
Collapse
Affiliation(s)
- Kate Karelina
- Department of Neuroscience, Group in Behavioral Neuroendocrinology, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Samuel Nicholson
- Department of Neuroscience, Group in Behavioral Neuroendocrinology, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | | |
Collapse
|
104
|
Intestinal dysbiosis and permeability: the yin and yang in alcohol dependence and alcoholic liver disease. Clin Sci (Lond) 2018; 132:199-212. [PMID: 29352076 DOI: 10.1042/cs20171055] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/07/2017] [Accepted: 12/19/2017] [Indexed: 02/07/2023]
Abstract
Alcohol dependence and alcoholic liver disease represent a major public health problem with substantial morbidity and mortality. By yet incompletely understood mechanisms, chronic alcohol abuse is associated with increased intestinal permeability and alterations of the gut microbiota composition, allowing bacterial components, bacteria, and metabolites to reach the portal and the systemic circulation. These gut-derived bacterial products are recognized by immune cells circulating in the blood or residing in remote organs such as the liver leading to the release of pro-inflammatory cytokines which are considered important mediators of the liver-gut-brain communication. Although circulating cytokines are likely not the sole factors involved, they can induce liver inflammation/damage and reach the central nervous system where they favor neuroinflammation which is associated with change in mood, cognition, and drinking behavior. In this review, the authors focus on the current evidence describing the changes that occur in the intestinal microbiota with chronic alcohol consumption in conjunction with intestinal barrier breakdown and inflammatory changes sustaining the concept of a gut-liver-brain axis in the pathophysiology of alcohol dependence and alcoholic liver disease.
Collapse
|
105
|
Flores-Bastías O, Karahanian E. Neuroinflammation produced by heavy alcohol intake is due to loops of interactions between Toll-like 4 and TNF receptors, peroxisome proliferator-activated receptors and the central melanocortin system: A novel hypothesis and new therapeutic avenues. Neuropharmacology 2018; 128:401-407. [DOI: 10.1016/j.neuropharm.2017.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 10/26/2017] [Accepted: 11/03/2017] [Indexed: 02/06/2023]
|
106
|
Lainiola M, Linden AM. Alcohol intake in two different mouse drinking models after recovery from the lipopolysaccharide-induced sickness reaction. Alcohol 2017; 65:1-10. [PMID: 29084623 DOI: 10.1016/j.alcohol.2017.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/19/2017] [Accepted: 06/12/2017] [Indexed: 12/19/2022]
Abstract
Neuroinflammation may play an important role in the development of alcohol addiction. Recent preclinical reports suggest that enhanced innate immune system signaling increases consumption of alcohol. Our aim was to study whether consequences of lipopolysaccharide (LPS)-induced sickness reaction increase long-term alcohol intake. Adult male C57BL/6J mice, housed in individually ventilated cages, were injected with LPS intraperitoneally (i.p.) and allowed to recover from an acute sickness reaction for 1 week before analysis of their alcohol intake in two different drinking models. Effects of LPS challenge were tested in a continuous two-bottle free choice test with increasing concentrations of alcohol and in a drinking in the dark (DID) binge model. In addition, the effect of repeatedly administered LPS during abstinence periods between binge drinking was analyzed in the DID model. In addition, the DID model was used to study the effects of the microglia inhibitor minocycline (50 mg/kg/day, 4 days) and purinergic P2X7 receptor antagonist Brilliant Blue G (75 mg/kg/day, 7 days) on alcohol intake. In contrast to previous findings, pretreatment with a 1-mg/kg dose of LPS did not significantly increase ethanol consumption in the continuous two-bottle choice test. As a novel finding, we report that increasing the LPS dose to 1.5 mg/kg reduced consumption of 18 and 21% (v/v) ethanol. In the DID model, pretreatment with LPS (0.2-1.5 mg/kg) did not significantly alter 15% or 20% ethanol consumption. Neither did repeated LPS injections affect binge alcohol drinking. Minocycline reduced alcohol, but also water, intake regardless of LPS pretreatment. No data on effects of P2X7 antagonists on alcohol consumption have been previously published; therefore, we report here that subchronic Brilliant Blue G had no effect on alcohol intake in the DID model. As a conclusion, further studies are needed to validate this LPS model of the interaction between immune system activation and alcohol consumption.
Collapse
|
107
|
Dakic V, Minardi Nascimento J, Costa Sartore R, Maciel RDM, de Araujo DB, Ribeiro S, Martins-de-Souza D, Rehen SK. Short term changes in the proteome of human cerebral organoids induced by 5-MeO-DMT. Sci Rep 2017; 7:12863. [PMID: 28993683 PMCID: PMC5634411 DOI: 10.1038/s41598-017-12779-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/14/2017] [Indexed: 12/28/2022] Open
Abstract
Dimethyltryptamines are entheogenic serotonin-like molecules present in traditional Amerindian medicine recently associated with cognitive gains, antidepressant effects, and changes in brain areas related to attention. Legal restrictions and the lack of adequate experimental models have limited the understanding of how such substances impact human brain metabolism. Here we used shotgun mass spectrometry to explore proteomic differences induced by 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) on human cerebral organoids. Out of the 6,728 identified proteins, 934 were found differentially expressed in 5-MeO-DMT-treated cerebral organoids. In silico analysis reinforced previously reported anti-inflammatory actions of 5-MeO-DMT and revealed modulatory effects on proteins associated with long-term potentiation, the formation of dendritic spines, including those involved in cellular protrusion formation, microtubule dynamics, and cytoskeletal reorganization. Our data offer the first insight about molecular alterations caused by 5-MeO-DMT in human cerebral organoids.
Collapse
Affiliation(s)
- Vanja Dakic
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Minardi Nascimento
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.,Laboratory of Neuroproteomics, Institute of Biology, Department of Biochemistry and Tissue Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Rafaela Costa Sartore
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Sidarta Ribeiro
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Institute of Biology, Department of Biochemistry and Tissue Biology, University of Campinas (UNICAMP), Campinas, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico, Sao Paulo, Brazil
| | - Stevens K Rehen
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil. .,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
108
|
Smith BL, Reyes TM. Offspring neuroimmune consequences of maternal malnutrition: Potential mechanism for behavioral impairments that underlie metabolic and neurodevelopmental disorders. Front Neuroendocrinol 2017; 47:109-122. [PMID: 28736323 PMCID: PMC8600507 DOI: 10.1016/j.yfrne.2017.07.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 07/19/2017] [Accepted: 07/20/2017] [Indexed: 12/18/2022]
Abstract
Maternal malnutrition significantly increases offspring risk for both metabolic and neurodevelopmental disorders. Animal models of maternal malnutrition have identified behavioral changes in the adult offspring related to executive function and reward processing. Together, these changes in executive and reward-based behaviors likely contribute to the etiology of both metabolic and neurodevelopmental disorders associated with maternal malnutrition. Concomitant with the behavioral effects, maternal malnutrition alters offspring expression of reward-related molecules and inflammatory signals in brain pathways that control executive function and reward. Neuroimmune pathways and microglial interactions in these specific brain circuits, either in early development or later in adulthood, could directly contribute to the maternal malnutrition-induced behavioral phenotypes. Understanding these mechanisms will help advance treatment strategies for metabolic and neurodevelopmental disorders, especially noninvasive dietary supplementation interventions.
Collapse
Affiliation(s)
- B L Smith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati OH, USA
| | - T M Reyes
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati OH, USA.
| |
Collapse
|
109
|
Terasaki LS, Schwarz JM. Impact of Prenatal and Subsequent Adult Alcohol Exposure on Pro-Inflammatory Cytokine Expression in Brain Regions Necessary for Simple Recognition Memory. Brain Sci 2017; 7:brainsci7100125. [PMID: 28973966 PMCID: PMC5664052 DOI: 10.3390/brainsci7100125] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/12/2017] [Accepted: 09/29/2017] [Indexed: 11/16/2022] Open
Abstract
Microglia, the immune cells of the brain, are important and necessary for appropriate neural development; however, activation of microglia, concomitant with increased levels of secreted immune molecules during brain development, can leave the brain susceptible to certain long-term changes in immune function associated with neurological and developmental disorders. One mechanism by which microglia can be activated is via alcohol exposure. We sought to investigate if low levels of prenatal alcohol exposure can alter the neuroimmune response to a subsequent acute dose of alcohol in adulthood. We also used the novel object location and recognition memory tasks to determine whether there are cognitive deficits associated with low prenatal alcohol exposure and subsequent adulthood alcohol exposure. We found that adult rats exposed to an acute binge-like level of alcohol, regardless of gestational alcohol exposure, have a robust increase in the expression of Interleukin (IL)-6 within the brain, and a significant decrease in the expression of IL-1β and CD11b. Rats exposed to alcohol during gestation, adulthood, or at both time points exhibited impaired cognitive performance in the cognitive tasks. These results indicate that both low-level prenatal alcohol exposure and even acute alcohol exposure in adulthood can significantly impact neuroimmune and associated cognitive function.
Collapse
Affiliation(s)
- Laurne S Terasaki
- Psychological and Brain Sciences, University of Delaware, 108 Wolf Hall, Newark, DE 19716, USA.
| | - Jaclyn M Schwarz
- Psychological and Brain Sciences, University of Delaware, 108 Wolf Hall, Newark, DE 19716, USA.
| |
Collapse
|
110
|
Karlsson C, Schank JR, Rehman F, Stojakovic A, Björk K, Barbier E, Solomon M, Tapocik J, Engblom D, Thorsell A, Heilig M. Proinflammatory signaling regulates voluntary alcohol intake and stress-induced consumption after exposure to social defeat stress in mice. Addict Biol 2017; 22:1279-1288. [PMID: 27273552 DOI: 10.1111/adb.12416] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 03/22/2016] [Accepted: 05/10/2016] [Indexed: 01/23/2023]
Abstract
Proinflammatory activity has been postulated to play a role in addictive processes and stress responses, but the underlying mechanisms remain largely unknown. Here, we examined the role of interleukin 1 (IL-1) and tumor necrosis factor-α (TNF-α) in regulation of voluntary alcohol consumption, alcohol reward and stress-induced drinking. Mice with a deletion of the IL-1 receptor I gene (IL-1RI KO) exhibited modestly decreased alcohol consumption. However, IL-1RI deletion affected neither the rewarding properties of alcohol, measured by conditioned place preference (CPP), nor stress-induced drinking induced by social defeat stress. TNF-α signaling can compensate for phenotypic consequences of IL1-RI deletion. We therefore hypothesized that double deletion of both IL-1RI and TNF-1 receptors (TNF-1R) may reveal the role of these pathways in regulation of alcohol intake. Double KOs consumed significantly less alcohol than control mice over a range of alcohol concentrations. The combined deletion of TNF-1R and IL-1RI did not influence alcohol reward, but did prevent increased alcohol consumption resulting from exposure to repeated bouts of social defeat stress. Taken together, these data indicate that IL-1RI and TNF-1R contribute to regulation of stress-induced, negatively reinforced drinking perhaps through overlapping signaling events downstream of these receptors, while leaving rewarding properties of alcohol largely unaffected.
Collapse
Affiliation(s)
- Camilla Karlsson
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Jesse R. Schank
- Department of Physiology and Pharmacology; University of Georgia; Athens GA
| | - Faazal Rehman
- Laboratory of Clinical and Translational Studies; National Institute of Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH); Bethesda MD USA
| | - Andrea Stojakovic
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Karl Björk
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Estelle Barbier
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Matthew Solomon
- Laboratory of Clinical and Translational Studies; National Institute of Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH); Bethesda MD USA
| | - Jenica Tapocik
- Laboratory of Clinical and Translational Studies; National Institute of Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH); Bethesda MD USA
| | - David Engblom
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Annika Thorsell
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Markus Heilig
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| |
Collapse
|
111
|
de Oliveira BM, Telles TM, Lomba LA, Correia D, Zampronio AR. Effects of binge-like ethanol exposure during adolescence on the hyperalgesia observed during sickness syndrome in rats. Pharmacol Biochem Behav 2017; 160:63-69. [DOI: 10.1016/j.pbb.2017.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/05/2017] [Accepted: 08/10/2017] [Indexed: 01/05/2023]
|
112
|
Cosa A, Moreno A, Pacheco-Torres J, Ciccocioppo R, Hyytiä P, Sommer WH, Moratal D, Canals S. Multi-modal MRI classifiers identify excessive alcohol consumption and treatment effects in the brain. Addict Biol 2017; 22:1459-1472. [PMID: 27273582 DOI: 10.1111/adb.12418] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 04/22/2016] [Accepted: 05/13/2016] [Indexed: 12/14/2022]
Abstract
Robust neuroimaging markers of neuropsychiatric disorders have proven difficult to obtain. In alcohol use disorders, profound brain structural deficits can be found in severe alcoholic patients, but the heterogeneity of unimodal MRI measurements has so far precluded the identification of selective biomarkers, especially for early diagnosis. In the present work we used a combination of multiple MRI modalities to provide comprehensive and insightful descriptions of brain tissue microstructure. We performed a longitudinal experiment using Marchigian-Sardinian (msP) rats, an established model of chronic excessive alcohol consumption, and acquired multi-modal images before and after 1 month of alcohol consumption (6.8 ± 1.4 g/kg/day, mean ± SD), as well as after 1 week of abstinence with or without concomitant treatment with the antirelapse opioid antagonist naltrexone (2.5 mg/kg/day). We found remarkable sensitivity and selectivity to accurately classify brains affected by alcohol even after the relative short exposure period. One month drinking was enough to imprint a highly specific signature of alcohol consumption. Brain alterations were regionally specific and affected both gray and white matter and persisted into the early abstinence state without any detectable recovery. Interestingly, naltrexone treatment during early abstinence resulted in subtle brain changes that could be distinguished from non-treated abstinent brains, suggesting the existence of an intermediate state associated with brain recovery from alcohol exposure induced by medication. The presented framework is a promising tool for the development of biomarkers for clinical diagnosis of alcohol use disorders, with capacity to further inform about its progression and response to treatment.
Collapse
Affiliation(s)
- Alejandro Cosa
- Instituto de Neurociencias; Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández; Sant Joan d'Alacant Spain
- Center for Biomaterials and Tissue Engineering; Universitat Politècnica de València; Valencia Spain
| | - Andrea Moreno
- Instituto de Neurociencias; Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández; Sant Joan d'Alacant Spain
| | - Jesús Pacheco-Torres
- Instituto de Neurociencias; Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández; Sant Joan d'Alacant Spain
| | | | - Petri Hyytiä
- Department of Pharmacology, Faculty of Medicine; University of Helsinki; Helsinki Finland
| | - Wolfgang H. Sommer
- Department of Psychopharmacology, Central Institute of Mental Health; University of Heidelberg; Mannheim Germany
| | - David Moratal
- Center for Biomaterials and Tissue Engineering; Universitat Politècnica de València; Valencia Spain
| | - Santiago Canals
- Instituto de Neurociencias; Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández; Sant Joan d'Alacant Spain
| |
Collapse
|
113
|
Neupane SP, Bramness JG, Lien L. Comorbid post-traumatic stress disorder in alcohol use disorder: relationships to demography, drinking and neuroimmune profile. BMC Psychiatry 2017; 17:312. [PMID: 28851339 PMCID: PMC5576315 DOI: 10.1186/s12888-017-1479-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 08/22/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND This study examined how alcohol use disorder (AUD) patients with post-traumatic stress disorder (PTSD) differed from those without PTSD in terms of demography, drinking patterns and C-reactive protein, inflammatory cytokines, tryptophan metabolism parameters, and brain-derived neurotrophic factor (BDNF). METHODS A consecutive sample (N = 187) of treatment-receiving AUD individuals were recruited from Nepalese facilities. They underwent fully structured psychiatric interviews. Serum levels of inflammatory cytokines [interleukin (IL)-6, IL-1 Receptor antagonist (IL-1Ra), IL-10, tumor necrosis factor-alpha (TNF-α), and interferon-gamma (IFN-γ)] were determined by a multiplex assay, kynurenine and tryptophan levels by high-performance liquid chromatography, and BDNF by enzyme-linked immunosorbent assay (ELISA). RESULTS The prevalence of exposure to severe trauma and PTSD was 74% and 17%, respectively. PTSD comorbidity was not associated with age, gender, or socioeconomic status, but with co-occurring major depression, history of attempted suicide, earlier peak of drinking problems, higher drinking quantity and withdrawal symptoms, experiencing alcoholic blackouts, and drinking problems among parents. None of the assessed neuroimmune parameters was related to comorbid PTSD. CONCLUSIONS The findings support routine trauma screening in AUD treatment samples and screening for risky drinking in trauma populations to help guide interventions. The expected aberrations in neuroimmune functioning may not be found when examined in a sample with multiple psychiatric morbidities.
Collapse
Affiliation(s)
- Sudan Prasad Neupane
- Norwegian National Advisory Unit on Concurrent Substance Abuse and Mental Health Disorders, Innlandet Hospital Trust, Post box 104, 2381, Brumunddal, Norway. .,Norwegian Centre for Addiction Research (SERAF), University of Oslo, Oslo, Norway.
| | - Jørgen G. Bramness
- grid.412929.5Norwegian National Advisory Unit on Concurrent Substance Abuse and Mental Health Disorders, Innlandet Hospital Trust, Post box 104, 2381 Brumunddal, Norway
| | - Lars Lien
- grid.412929.5Norwegian National Advisory Unit on Concurrent Substance Abuse and Mental Health Disorders, Innlandet Hospital Trust, Post box 104, 2381 Brumunddal, Norway ,grid.477237.2Department of Public Health, Hedmark University College, Elverum, Norway
| |
Collapse
|
114
|
Nennig SE, Schank JR. The Role of NFkB in Drug Addiction: Beyond Inflammation. Alcohol Alcohol 2017; 52:172-179. [PMID: 28043969 DOI: 10.1093/alcalc/agw098] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Indexed: 12/19/2022] Open
Abstract
Aims Nuclear factor kappa light chain enhancer of activated B cells (NFkB) is a ubiquitous transcription factor well known for its role in the innate immune response. As such, NFkB is a transcriptional activator of inflammatory mediators such as cytokines. It has recently been demonstrated that alcohol and other drugs of abuse can induce NFkB activity and cytokine expression in the brain. A number of reviews have been published highlighting this effect of alcohol, and have linked increased NFkB function to neuroimmune-stimulated toxicity. However, in this review we focus on the potentially non-immune functions of NFkB as possible links between NFkB and addiction. Methods An extensive review of the literature via Pubmed searches was used to assess the current state of the field. Results NFkB can induce the expression of a diverse set of gene targets besides inflammatory mediators, some of which are involved in addictive processes, such as opioid receptors and neuropeptides. NFkB mediates complex behaviors including learning and memory, stress responses, anhedonia and drug reward, processes that may lie outside the role of NFkB in the classic neuroimmune response. Conclusions Future studies should focus on these non-immune functions of NFkB signaling and their association with addiction-related processes.
Collapse
Affiliation(s)
- S E Nennig
- Department of Physiology and Pharmacology, University of Georgia, 501 D.W. Brooks Drive, Athens, GA 30602, USA
| | - J R Schank
- Department of Physiology and Pharmacology, University of Georgia, 501 D.W. Brooks Drive, Athens, GA 30602, USA
| |
Collapse
|
115
|
Increased expression of M1 and M2 phenotypic markers in isolated microglia after four-day binge alcohol exposure in male rats. Alcohol 2017; 62:29-40. [PMID: 28755749 DOI: 10.1016/j.alcohol.2017.02.175] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/30/2017] [Accepted: 02/06/2017] [Indexed: 01/21/2023]
Abstract
Microglia activation and neuroinflammation are common features of neurodegenerative conditions, including alcohol use disorders (AUDs). When activated, microglia span a continuum of diverse phenotypes ranging from classically activated, pro-inflammatory (M1) microglia/macrophages to alternatively activated, growth-promoting (M2) microglia/macrophages. Identifying microglia phenotypes is critical for understanding the role of microglia in the pathogenesis of AUDs. Therefore, male rats were gavaged with 25% (w/v) ethanol or isocaloric control diet every 8 h for 4 days and sacrificed at 0, 2, 4, and 7 days after alcohol exposure (e.g., T0, T2, etc.). Microglia were isolated from hippocampus and entorhinal cortices by Percoll density gradient centrifugation. Cells were labeled with microglia surface antigens and analyzed by flow cytometry. Consistent with prior studies, isolated cells yielded a highly enriched population of brain macrophages/microglia (>95% pure), evidenced by staining for the macrophage/microglia antigen CD11b. Polarization states of CD11b+CD45low microglia were evaluated by expression of M1 surface markers, major histocompatibility complex (MHC) II, CD32, CD86, and M2 surface marker, CD206 (mannose receptor). Ethanol-treated animals begin to show increased expression of M1 and M2 markers at T0 (p = n.s.), with significant changes at the T2 time point. At T2, expression of M1 markers, MHC-II, CD86, and CD32 were increased (p < 0.05) in hippocampus and entorhinal cortices, while M2 marker, CD206, was increased significantly only in entorhinal cortices (p < 0.05). All effects resolved to control levels by T4. In summary, four-day binge alcohol exposure produces a transient increase in both M1 (MHC-II, CD32, and CD86) and M2 (CD206) populations of microglia isolated from the entorhinal cortex and hippocampus. Thus, these findings that both pro-inflammatory and potentially beneficial, recovery-promoting microglia phenotypes can be observed after a damaging exposure of alcohol are critically important to our understanding of the role of microglia in the pathogenesis of AUDs.
Collapse
|
116
|
Cippitelli A, Domi E, Ubaldi M, Douglas JC, Li HW, Demopulos G, Gaitanaris G, Roberto M, Drew PD, Kane CJM, Ciccocioppo R. Protection against alcohol-induced neuronal and cognitive damage by the PPARγ receptor agonist pioglitazone. Brain Behav Immun 2017; 64:320-329. [PMID: 28167117 PMCID: PMC5482782 DOI: 10.1016/j.bbi.2017.02.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/25/2017] [Accepted: 02/01/2017] [Indexed: 01/08/2023] Open
Abstract
Binge alcohol drinking has emerged as a typical phenomenon in young people. This pattern of drinking, repeatedly leading to extremely high blood and brain alcohol levels and intoxication is associated with severe risks of neurodegeneration and cognitive damage. Mechanisms involved in excitotoxicity and neuroinflammation are pivotal elements in alcohol-induced neurotoxicity. Evidence has demonstrated that PPARγ receptor activation shows anti-inflammatory and neuroprotective properties. Here we examine whether treatment with the PPARγ agonist pioglitazone is beneficial in counteracting neurodegeneration, neuroinflammation and cognitive damage produced by binge alcohol intoxication. Adult Wistar rats were subjected to a 4-day binge intoxication procedure, which is commonly used to model excessive alcohol consumption in humans. Across the 4-day period, pioglitazone (0, 30, 60mg/kg) was administered orally twice daily at 12-h intervals. Degenerative cells were detected by fluoro-jade B (FJ-B) immunostaining in brain regions where expression of pro-inflammatory cytokines was also determined. The effects of pioglitazone on cognitive function were assessed in an operant reversal learning task and the Morris water maze task. Binge alcohol exposure produced selective neuronal degeneration in the hippocampal dentate gyrus and the adjacent entorhinal cortex. Pioglitazone reduced FJ-B positive cells in both regions and prevented alcohol-induced expression of pro-inflammatory cytokines. Pioglitazone also rescued alcohol-impaired reversal learning in the operant task and spatial learning deficits in the Morris water maze. These findings demonstrate that activation of PPARγ protects against neuronal and cognitive degeneration elicited by binge alcohol exposure. The protective effect of PPARγ agonist appears to be linked to inhibition of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Andrea Cippitelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino (MC) 62032, Italy
| | - Esi Domi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino (MC) 62032, Italy
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino (MC) 62032, Italy
| | - James C. Douglas
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Hong Wu Li
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino (MC) 62032, Italy
| | | | | | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Paul D. Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Cynthia J. M. Kane
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino (MC) 62032, Italy.
| |
Collapse
|
117
|
Weil ZM, Karelina K. Traumatic Brain Injuries during Development: Implications for Alcohol Abuse. Front Behav Neurosci 2017; 11:135. [PMID: 28775682 PMCID: PMC5517445 DOI: 10.3389/fnbeh.2017.00135] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/07/2017] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injuries are strongly related to alcohol intoxication as by some estimates half or more of all brain injuries involve at least one intoxicated individual. Additionally, there is mounting evidence that traumatic brain injuries can themselves serve as independent risk factors for the development of alcohol use disorders, particularly when injury occurs during juvenile or adolescent development. Here, we will review the epidemiological and experimental evidence for this phenomenon and discuss potential psychosocial mediators including attenuation of negative affect and impaired decision making as well as neurochemical mediators including disruption in the glutamatergic, GABAergic, and dopaminergic signaling pathways and increases in inflammation.
Collapse
Affiliation(s)
- Zachary M Weil
- Behavioral Neuroendocrinology Group, Department of Neuroscience, Center for Brain and Spinal Cord Repair, Ohio State University Wexner Medical CenterColumbus, OH, United States
| | - Kate Karelina
- Behavioral Neuroendocrinology Group, Department of Neuroscience, Center for Brain and Spinal Cord Repair, Ohio State University Wexner Medical CenterColumbus, OH, United States
| |
Collapse
|
118
|
Schunck RVA, Macedo IC, Laste G, de Souza A, Valle MTC, Salomón JLO, Nunes EA, Campos ACW, Gnoatto SCB, Bergold AM, Konrath EL, Dallegrave E, Arbo MD, Torres ILS, Leal MB. Standardized Passiflora incarnata L. Extract Reverts the Analgesia Induced by Alcohol Withdrawal in Rats. Phytother Res 2017; 31:1199-1208. [PMID: 28568647 DOI: 10.1002/ptr.5839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 04/14/2017] [Accepted: 05/03/2017] [Indexed: 12/13/2022]
Abstract
Passiflora incarnata L. (Passifloraceae) has been traditionally used for treatment of anxiety, insomnia, drug addiction, mild infections, and pain. The aim of this study was to investigate the effect of a commercial extract of P. incarnata in the analgesia induced by alcohol withdrawal syndrome in rats. In addition, brain-derived neurotrophic factor and interleukin-10 levels were evaluated in prefrontal cortex, brainstem, and hippocampus. Male adult rats received by oral gavage: (1: water group) water for 19 days, 1 day interval and water (8 days); (2: P. incarnata group) water for 19 days, 1 day interval and P. incarnata 200 mg/kg (8 days); (3: alcohol withdrawal group) alcohol for 19 days, 1 day interval and water (8 days); and (4: P. incarnata in alcohol withdrawal) alcohol for 19 days, 1 day interval and P. incarnata 200 mg/kg (8 days). The tail-flick and hot plate tests were used as nociceptive response measures. Confirming previous study of our group, it was showed that alcohol-treated groups presented an increase in the nociceptive thresholds after alcohol withdrawal, which was reverted by P. incarnata, measured by the hot plate test. Besides, alcohol treatment increased brain-derived neurotrophic factor and interleukin-10 levels in prefrontal cortex, which was not reverted by P. incarnata. Considering these results, the P. incarnata treatment might be a potential therapy in the alcohol withdrawal syndrome. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Rebeca Vargas Antunes Schunck
- Programa de Pós-Graduação em Ciências Biológicas - Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/107, 90050-170, Porto Alegre, RS, Brazil.,Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/202, 90050-170, Porto Alegre, RS, Brazil
| | - Isabel Cristina Macedo
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, 90050-170, Porto Alegre, RS, Brazil
| | - Gabriela Laste
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, 90050-170, Porto Alegre, RS, Brazil
| | - Andressa de Souza
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, 90050-170, Porto Alegre, RS, Brazil
| | - Marina Tuerlinckx Costa Valle
- Programa de Pós-Graduação em Ciências Biológicas - Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/107, 90050-170, Porto Alegre, RS, Brazil.,Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/202, 90050-170, Porto Alegre, RS, Brazil
| | - Janaína L O Salomón
- Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/202, 90050-170, Porto Alegre, RS, Brazil
| | - Ellen Almeida Nunes
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, 90050-170, Porto Alegre, RS, Brazil
| | - Andreia Cristina Wildner Campos
- Departamento de Produção de Matéria-Prima, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Simone Cristina Baggio Gnoatto
- Departamento de Produção de Matéria-Prima, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Ana Maria Bergold
- Departamento de Produção de Matéria-Prima, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Eduardo L Konrath
- Departamento de Produção de Matéria-Prima, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Eliane Dallegrave
- Departamento de Farmacociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245, 90050-170, Porto Alegre, RS, Brazil
| | - Marcelo Dutra Arbo
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, 90050-000, Porto Alegre, RS, Brazil
| | - Iraci L S Torres
- Programa de Pós-Graduação em Ciências Biológicas - Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/107, 90050-170, Porto Alegre, RS, Brazil.,Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, 90050-170, Porto Alegre, RS, Brazil
| | - Mirna Bainy Leal
- Programa de Pós-Graduação em Ciências Biológicas - Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/107, 90050-170, Porto Alegre, RS, Brazil.,Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/202, 90050-170, Porto Alegre, RS, Brazil
| |
Collapse
|
119
|
Vallender EJ, Goswami DB, Shinday NM, Westmoreland SV, Yao WD, Rowlett JK. Transcriptomic profiling of the ventral tegmental area and nucleus accumbens in rhesus macaques following long-term cocaine self-administration. Drug Alcohol Depend 2017; 175:9-23. [PMID: 28376414 PMCID: PMC5693237 DOI: 10.1016/j.drugalcdep.2017.01.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 01/18/2017] [Accepted: 01/18/2017] [Indexed: 01/23/2023]
Abstract
BACKGROUND The behavioral consequences associated with addiction are thought to arise from drug-induced neuroadaptation. The mesolimbic system plays an important initial role in this process, and while the dopaminergic system specifically has been strongly interrogated, a complete understanding of the broad transcriptomic effects associated with cocaine use remains elusive. METHODS Using next generation sequencing approaches, we performed a comprehensive evaluation of gene expression differences in the ventral tegmental area and nucleus accumbens of rhesus macaques that had self-administered cocaine for roughly 100days and saline-yoked controls. During self-administration, the monkeys increased daily consumption of cocaine until almost the maximum number of injections were taken within the first 15min of the one hour session for a total intake of 3mg/kg/day. RESULTS We confirm the centrality of dopaminergic differences in the ventral tegmental area, but in the nucleus accumbens we see the strongest evidence for an inflammatory response and large scale chromatin remodeling. CONCLUSIONS These findings suggest an expanded understanding of the pathology of cocaine addiction with the potential to lead to the development of alternative treatment strategies.
Collapse
Affiliation(s)
- Eric J. Vallender
- Harvard Medical School, New England Primate Research Center, Southborough, MA 01772,University of Mississippi Medical Center, Jackson, MS 39216,Tulane National Primate Research Center, Covington, LA 70433
| | - Dharmendra B. Goswami
- Harvard Medical School, New England Primate Research Center, Southborough, MA 01772,Boston University, Boston, MA 02118
| | - Nina M. Shinday
- Harvard Medical School, New England Primate Research Center, Southborough, MA 01772,University of Massachusetts-Amherst, Amherst, MA 01003
| | | | - Wei-Dong Yao
- Harvard Medical School, New England Primate Research Center, Southborough, MA 01772,SUNY Upstate Medical University, Syracuse, NY 13210
| | - James K. Rowlett
- Harvard Medical School, New England Primate Research Center, Southborough, MA 01772,University of Mississippi Medical Center, Jackson, MS 39216,Tulane National Primate Research Center, Covington, LA 70433,University of Massachusetts-Amherst, Amherst, MA 01003
| |
Collapse
|
120
|
Crews FT, Walter TJ, Coleman LG, Vetreno RP. Toll-like receptor signaling and stages of addiction. Psychopharmacology (Berl) 2017; 234:1483-1498. [PMID: 28210782 PMCID: PMC5420377 DOI: 10.1007/s00213-017-4560-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/03/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Athina Markou and her colleagues discovered persistent changes in adult behavior following adolescent exposure to ethanol or nicotine consistent with increased risk for developing addiction. Building on Dr. Markou's important work and that of others in the field, researchers at the Bowles Center for Alcohol Studies have found that persistent changes in behavior following adolescent stress or alcohol exposure may be linked to induction of immune signaling in brain. AIM This study aims to illuminate the critical interrelationship of the innate immune system (e.g., toll-like receptors [TLRs], high-mobility group box 1 [HMGB1]) in the neurobiology of addiction. METHOD This study reviews the relevant research regarding the relationship between the innate immune system and addiction. CONCLUSION Emerging evidence indicates that TLRs in brain, particularly those on microglia, respond to endogenous innate immune agonists such as HMGB1 and microRNAs (miRNAs). Multiple TLRs, HMGB1, and miRNAs are induced in the brain by stress, alcohol, and other drugs of abuse and are increased in the postmortem human alcoholic brain. Enhanced TLR-innate immune signaling in brain leads to epigenetic modifications, alterations in synaptic plasticity, and loss of neuronal cell populations, which contribute to cognitive and emotive dysfunctions. Addiction involves progressive stages of drug binges and intoxication, withdrawal-negative affect, and ultimately compulsive drug use and abuse. Toll-like receptor signaling within cortical-limbic circuits is modified by alcohol and stress in a manner consistent with promoting progression through the stages of addiction.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - T Jordan Walter
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
121
|
Zerdazi EH, Oliveira J, Vorspan F, Bennabi M, Jamain S, Etain B, Leboyer M, Tamouza R, Bellivier F. TLR4 gene polymorphism associated with lifetime cigarette smoking in bipolar disorder. J Neuroimmunol 2017; 305:96-101. [DOI: 10.1016/j.jneuroim.2017.01.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 02/06/2023]
|
122
|
Merkel SF, Cannella LA, Razmpour R, Lutton E, Raghupathi R, Rawls SM, Ramirez SH. Factors affecting increased risk for substance use disorders following traumatic brain injury: What we can learn from animal models. Neurosci Biobehav Rev 2017; 77:209-218. [PMID: 28359860 DOI: 10.1016/j.neubiorev.2017.03.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/06/2017] [Accepted: 03/26/2017] [Indexed: 11/17/2022]
Abstract
Recent studies have helped identify multiple factors affecting increased risk for substance use disorders (SUDs) following traumatic brain injury (TBI). These factors include age at the time of injury, repetitive injury and TBI severity, neurocircuits, neurotransmitter systems, neuroinflammation, and sex differences. This review will address each of these factors by discussing 1) the clinical and preclinical data identifying patient populations at greatest risk for SUDs post-TBI, 2) TBI-related neuropathology in discrete brain regions heavily implicated in SUDs, and 3) the effects of TBI on molecular mechanisms that may drive substance abuse behavior, like dopaminergic and glutamatergic transmission or neuroimmune signaling in mesolimbic regions of the brain. Although these studies have laid the groundwork for identifying factors that affect risk of SUDs post-TBI, additional studies are required. Notably, preclinical models have been shown to recapitulate many of the behavioral, cellular, and neurochemical features of SUDs and TBI. Therefore, these models are well suited for answering important questions that remain in future investigations.
Collapse
Affiliation(s)
- Steven F Merkel
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Lee Anne Cannella
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Roshanak Razmpour
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Evan Lutton
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Ramesh Raghupathi
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Scott M Rawls
- Department of Pharmacology, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Servio H Ramirez
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; Shriners Hospitals Pediatric Research Center, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
123
|
Warden AS, Mayfield RD. Gene expression profiling in the human alcoholic brain. Neuropharmacology 2017; 122:161-174. [PMID: 28254370 DOI: 10.1016/j.neuropharm.2017.02.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/13/2017] [Accepted: 02/17/2017] [Indexed: 01/12/2023]
Abstract
Long-term alcohol use causes widespread changes in gene expression in the human brain. Aberrant gene expression changes likely contribute to the progression from occasional alcohol use to alcohol use disorder (including alcohol dependence). Transcriptome studies have identified individual gene candidates that are linked to alcohol-dependence phenotypes. The use of bioinformatics techniques to examine expression datasets has provided novel systems-level approaches to transcriptome profiling in human postmortem brain. These analytical advances, along with recent developments in next-generation sequencing technology, have been instrumental in detecting both known and novel coding and non-coding RNAs, alternative splicing events, and cell-type specific changes that may contribute to alcohol-related pathologies. This review offers an integrated perspective on alcohol-responsive transcriptional changes in the human brain underlying the regulatory gene networks that contribute to alcohol dependence. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Anna S Warden
- Institute for Neuroscience, The University of Texas at Austin, 1 University Station, C7000, Austin, TX 78712, USA; Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, 2500 Speedway, A4800, Austin, TX 78712, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, 2500 Speedway, A4800, Austin, TX 78712, USA.
| |
Collapse
|
124
|
Baxter-Potter LN, Henricks AM, Berger AL, Bieniasz KV, Lugo JM, McLaughlin RJ. Alcohol vapor exposure differentially impacts mesocorticolimbic cytokine expression in a sex-, region-, and duration-specific manner. Neuroscience 2017; 346:238-246. [DOI: 10.1016/j.neuroscience.2017.01.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/22/2016] [Accepted: 01/09/2017] [Indexed: 01/12/2023]
|
125
|
Systemic inflammation enhances stimulant-induced striatal dopamine elevation. Transl Psychiatry 2017; 7:e1076. [PMID: 28350401 PMCID: PMC5404612 DOI: 10.1038/tp.2017.18] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/10/2016] [Accepted: 01/09/2017] [Indexed: 12/25/2022] Open
Abstract
Changes in the mesolimbic dopamine (DA) system are implicated in a range of neuropsychiatric conditions including addiction, depression and schizophrenia. Dysfunction of the neuroimmune system is often comorbid with such conditions and affects similar areas of the brain. The goal of this study was to use positron emission tomography with the dopamine D2 antagonist tracer, 11C-raclopride, to explore the effect of acute immune activation on striatal DA levels. DA transmission was modulated by an oral methylphenidate (MP) challenge in order to reliably elicit DA elevation. Elevation in DA concentration due to MP was estimated via change in 11C-raclopride binding potential from the baseline scan. Prior to the post-MP scan, subjects were pre-treated with either the immune activator lipopolysaccharide (LPS) or placebo (PBO) in a cross-over design. Immune activation was confirmed by measuring tumor necrosis factor alpha (TNFα), interleukin (IL)-6 and IL-8 concentration in plasma. Eight healthy subjects were scanned four times each to determine the MP-induced DA elevation under both LPS and PBO pre-treatment conditions. MP-induced DA elevation in the striatum was significantly greater (P<0.01) after LPS pre-treatment compared to PBO pre-treatment. Seven of eight subjects responded similarly. This effect was observed in the caudate and putamen (P<0.02), but was not present in ventral striatum. DA elevation induced by MP was significantly greater when subjects were pre-treated with LPS compared to PBO. The amplification of stimulant-induced DA signaling in the presence of systemic inflammation may have important implications for our understanding of addiction and other diseases of DA dysfunction.
Collapse
|
126
|
Telles TM, de Oliveira BM, Lomba LA, Leite-Avalca MG, Correia D, Zampronio AR. Effects of Binge-Like Ethanol Exposure During Adolescence on the Febrile Response in Rats. Alcohol Clin Exp Res 2017; 41:507-515. [DOI: 10.1111/acer.13333] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 12/26/2016] [Indexed: 01/26/2023]
Affiliation(s)
| | | | - Luis A. Lomba
- Department of Pharmacology; Federal University of Paraná; Curitiba PR Brazil
| | | | - Diego Correia
- Department of General Biology; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | | |
Collapse
|
127
|
Vetreno RP, Patel Y, Patel U, Walter TJ, Crews FT. Adolescent intermittent ethanol reduces serotonin expression in the adult raphe nucleus and upregulates innate immune expression that is prevented by exercise. Brain Behav Immun 2017; 60:333-345. [PMID: 27647531 PMCID: PMC5215774 DOI: 10.1016/j.bbi.2016.09.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/29/2016] [Accepted: 09/16/2016] [Indexed: 12/18/2022] Open
Abstract
Serotonergic neurons of the raphe nucleus regulate sleep, mood, endocrine function, and other processes that mature during adolescence. Alcohol abuse and binge drinking are common during human adolescence. We tested the novel hypothesis that adolescent intermittent ethanol exposure would alter the serotonergic system that would persist into adulthood. Using a Wistar rat model of adolescent intermittent ethanol (AIE; 5.0g/kg, i.g., 2-day on/2-day off from postnatal day [P]25 to P55), we found a loss of dorsal raphe nucleus (DRN) serotonin (5-HT)-immunoreactive (+IR) neurons that persisted from late adolescence (P56) into adulthood (P220). Hypothalamic and amygdalar DRN serotonergic projections were reduced following AIE. Tryptophan hydroxylase 2, the rate-limiting 5-HT synthesizing enzyme, and vesicular monoamine transporter 2, which packages 5-HT into synaptic vesicles, were also reduced in the young adult midbrain following AIE treatment. Adolescent intermittent ethanol treatment increased expression of phosphorylated (activated) NF-κB p65 as well as markers of microglial activation (i.e., Iba-1 and CD11b) in the adult DRN. Administration of lipopolysaccharide to mimic AIE-induced innate immune activation reduced 5-HT+IR and increased phosphorylated NF-κB p65+IR similar to AIE treatment. Voluntary exercise during adolescence through young adulthood blunted microglial marker and phosphorylated NF-κB p65+IR, and prevented the AIE-induced loss of 5-HT+IR neurons in the DRN. Together, these novel data reveal that AIE reduces 5-HT+IR neurons in the adult DRN, possibly through an innate immune mechanism, which might impact adult cognition, arousal, or reward sensitivity. Further, exercise prevents the deleterious effects of AIE on the serotonergic system.
Collapse
Affiliation(s)
- Ryan P. Vetreno
- Corresponding author: Ryan P. Vetreno, Ph.D., University of North Carolina at Chapel Hill, School of Medicine, Bowles Center for Alcohol Studies, CB #7178, 1021 Thurston-Bowles Building, Chapel Hill, NC 27599-7178, Tel: 1-919-966-0501, Fax: 1-919-966-5679,
| | | | | | | | | |
Collapse
|
128
|
The role of neuroimmune signaling in alcoholism. Neuropharmacology 2017; 122:56-73. [PMID: 28159648 DOI: 10.1016/j.neuropharm.2017.01.031] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/24/2017] [Accepted: 01/29/2017] [Indexed: 02/07/2023]
Abstract
Alcohol consumption and stress increase brain levels of known innate immune signaling molecules. Microglia, the innate immune cells of the brain, and neurons respond to alcohol, signaling through Toll-like receptors (TLRs), high-mobility group box 1 (HMGB1), miRNAs, pro-inflammatory cytokines and their associated receptors involved in signaling between microglia, other glia and neurons. Repeated cycles of alcohol and stress cause a progressive, persistent induction of HMGB1, miRNA and TLR receptors in brain that appear to underlie the progressive and persistent loss of behavioral control, increased impulsivity and anxiety, as well as craving, coupled with increasing ventral striatal responses that promote reward seeking behavior and increase risk of developing alcohol use disorders. Studies employing anti-oxidant, anti-inflammatory, anti-depressant, and innate immune antagonists further link innate immune gene expression to addiction-like behaviors. Innate immune molecules are novel targets for addiction and affective disorders therapies. This article is part of the Special Issue entitled "Alcoholism".
Collapse
|
129
|
Karelina K, Gaier KR, Prabhu M, Wenger V, Corrigan TED, Weil ZM. Binge ethanol in adulthood exacerbates negative outcomes following juvenile traumatic brain injury. Brain Behav Immun 2017; 60:304-311. [PMID: 27845195 DOI: 10.1016/j.bbi.2016.11.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/04/2016] [Accepted: 11/08/2016] [Indexed: 01/26/2023] Open
Abstract
Traumatic brain injuries (TBI) are a major public health problem with enormous costs in terms of health care dollars, lost productivity, and reduced quality of life. Alcohol is bidirectionally linked to TBI as many TBI patients are intoxicated at the time of their injury and we recently reported that, in accordance with human epidemiological data, animals injured during juvenile development self-administered significantly more alcohol as adults than did sham injured mice. There are also clinical data that drinking after TBI significantly reduces the efficacy of rehabilitation and leads to poorer long-term outcomes. In order to determine whether juvenile traumatic brain injury also increased the vulnerability of the brain to the toxic effects of high dose alcohol, mice were injured at 21days of age and then seven weeks later treated daily with binge-like levels of alcohol 5g/kg (by oral gavage) for ten days. Binge-like alcohol produced a greater degree of neuronal damage and neuroinflammation in mice that sustained a TBI. Further, mice that sustained a juvenile TBI exhibited mild learning and memory impairments in adulthood following binge alcohol and express a significant increase in hippocampal ectopic localization of newborn neurons. Taken together, these data provide strong evidence that a mild brain injury occurring early in life renders the brain highly vulnerable to the consequences of binge-like alcohol consumption.
Collapse
Affiliation(s)
- Kate Karelina
- Department of Neuroscience, Group in Behavioral Neuroendocrinology and Center for Brain and Spinal Cord Repair, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Kristopher R Gaier
- Department of Neuroscience, Group in Behavioral Neuroendocrinology and Center for Brain and Spinal Cord Repair, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Maya Prabhu
- Department of Neuroscience, Group in Behavioral Neuroendocrinology and Center for Brain and Spinal Cord Repair, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Vanessa Wenger
- Department of Neuroscience, Group in Behavioral Neuroendocrinology and Center for Brain and Spinal Cord Repair, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Timothy E D Corrigan
- Department of Neuroscience, Group in Behavioral Neuroendocrinology and Center for Brain and Spinal Cord Repair, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Zachary M Weil
- Department of Neuroscience, Group in Behavioral Neuroendocrinology and Center for Brain and Spinal Cord Repair, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
130
|
The link between inflammation, bugs, the intestine and the brain in alcohol dependence. Transl Psychiatry 2017; 7:e1048. [PMID: 28244981 PMCID: PMC5545644 DOI: 10.1038/tp.2017.15] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 01/02/2017] [Accepted: 01/09/2017] [Indexed: 02/08/2023] Open
Abstract
In recent years, some new processes have been proposed to explain how alcohol may influence behavior, psychological symptoms and alcohol seeking in alcohol-dependent subjects. In addition to its important effect on brain and neurotransmitters equilibrium, alcohol abuse also affects peripheral organs including the gut. By yet incompletely understood mechanisms, chronic alcohol abuse increases intestinal permeability and alters the composition of the gut microbiota, allowing bacterial components from the gut lumen to reach the systemic circulation. These gut-derived bacterial products are recognized by immune cells circulating in the blood or residing in target organs, which consequently synthesize and release pro-inflammatory cytokines. Circulating cytokines are considered important mediators of the gut-brain communication, as they can reach the central nervous system and induce neuroinflammation that is associated with change in mood, cognition and drinking behavior. These observations support the possibility that targeting the gut microbiota, by the use of probiotics or prebiotics, could restore the gut barrier function, reduce systemic inflammation and may have beneficial effect in treating alcohol dependence and in reducing alcohol relapse.
Collapse
|
131
|
Monnig MA. Immune activation and neuroinflammation in alcohol use and HIV infection: evidence for shared mechanisms. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2017; 43:7-23. [PMID: 27532935 PMCID: PMC5250549 DOI: 10.1080/00952990.2016.1211667] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 07/07/2016] [Accepted: 07/07/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND Emerging research points to innate immune mechanisms in the neuropathological and behavioral consequences of heavy alcohol use. Alcohol use is common among people living with HIV infection (PLWH), a chronic condition that carries its own set of long-term effects on brain and behavior. Notably, neurobiological and cognitive profiles associated with heavy alcohol use and HIV infection share several prominent features. This observation raises questions about interacting biological mechanisms as well as compounded impairment when HIV infection and heavy drinking co-occur. OBJECTIVE AND METHOD This narrative overview discusses peer-reviewed research on specific immune mechanisms of alcohol that exhibit apparent potential to compound the neurobiological and psychiatric sequelae of HIV infection. These include microbial translocation, systemic immune activation, blood-brain barrier compromise, microglial activation, and neuroinflammation. RESULTS Clinical and preclinical evidence supports overlapping mechanistic actions of HIV and alcohol use on peripheral and neural immune systems. In preclinical studies, innate immune signaling mediates many of the detrimental neurocognitive and behavioral effects of alcohol use. Neuropsychopharmacological research suggests potential for a feed-forward cycle in which heavy drinking induces innate immune signaling, which in turn stimulates subsequent alcohol use behavior. CONCLUSION Alcohol-induced immune activation and neuroinflammation are a serious health concern for PLWH. Future research to investigate specific immune effects of alcohol in the context of HIV infection has potential to identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Mollie A. Monnig
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI
| |
Collapse
|
132
|
Neupane SP. Neuroimmune Interface in the Comorbidity between Alcohol Use Disorder and Major Depression. Front Immunol 2016; 7:655. [PMID: 28082989 PMCID: PMC5186784 DOI: 10.3389/fimmu.2016.00655] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 12/15/2016] [Indexed: 12/12/2022] Open
Abstract
Bidirectional communication links operate between the brain and the body. Afferent immune-to-brain signals are capable of inducing changes in mood and behavior. Chronic heavy alcohol drinking, typical of alcohol use disorder (AUD), is one such factor that provokes an immune response in the periphery that, by means of circulatory cytokines and other neuroimmune mediators, ultimately causes alterations in the brain function. Alcohol can also directly impact the immune functions of microglia, the resident immune cells of the central nervous system (CNS). Several lines of research have established the contribution of specific inflammatory mediators in the development and progression of depressive illness. Much of the available evidence in this field stems from cross-sectional data on the immune interactions between isolated AUD and major depression (MD). Given their heterogeneity as disease entities with overlapping symptoms and shared neuroimmune correlates, it is no surprise that systemic and CNS inflammation could be a critical determinant of the frequent comorbidity between AUD and MD. This review presents a summary and analysis of the extant literature on neuroimmune interface in the AUD–MD comorbidity.
Collapse
Affiliation(s)
- Sudan Prasad Neupane
- Norwegian National Advisory Unit on Concurrent Substance Abuse and Mental Health Disorders, Innlandet Hospital Trust, Brumunddal, Norway; Norwegian Centre for Addiction Research (SERAF), University of Oslo, Oslo, Norway
| |
Collapse
|
133
|
Traumatic Brain Injury and Substance Related Disorder: A 10-Year Nationwide Cohort Study in Taiwan. Neural Plast 2016; 2016:8030676. [PMID: 27774322 PMCID: PMC5059606 DOI: 10.1155/2016/8030676] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023] Open
Abstract
Whether traumatic brain injury (TBI) is causally related to substance related disorder (SRD) is still debatable, especially in persons with no history of mental disorders at the time of injury. This study analyzed data in the Taiwan National Health Insurance Research Database for 19,109 patients aged ≥18 years who had been diagnosed with TBI during 2000-2010. An additional 19,109 randomly selected age and gender matched patients without TBI (1 : 1 ratio) were enrolled in the control group. The relationship between TBI and SRD was estimated with Cox proportional hazard regression models. During the follow-up period, SRD developed in 340 patients in the TBI group and in 118 patients in the control group. After controlling for covariates, the overall incidence of SRD was 3.62-fold higher in the TBI group compared to the control group. Additionally, patients in the severe TBI subgroup were 9.01 times more likely to have SRD compared to controls. Notably, patients in the TBI group were prone to alcohol related disorders. The data in this study indicate that TBI is significantly associated with the subsequent risk of SRD. Physicians treating patients with TBI should be alert to this association to prevent the occurrence of adverse events.
Collapse
|
134
|
Poland RS, Hahn Y, Knapp PE, Beardsley PM, Bowers MS. Ibudilast attenuates expression of behavioral sensitization to cocaine in male and female rats. Neuropharmacology 2016; 109:281-292. [PMID: 27343385 PMCID: PMC5404892 DOI: 10.1016/j.neuropharm.2016.06.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/25/2016] [Accepted: 06/21/2016] [Indexed: 01/12/2023]
Abstract
There are no FDA-approved pharmacotherapies for cocaine use disorder, indicating a need to identify novel reagents with therapeutic potential. Ibudilast is an anti-inflammatory glial attenuator and non-selective phosphodiesterase inhibitor currently undergoing clinical evaluations for methamphetamine, opiate, and alcohol abuse disorders. We previously showed that twice daily (b.i.d.) ibudilast reduces the development of methamphetamine sensitization in male mice. However, nothing is known about the ability of ibudilast to modulate the expression of sensitization that occurs after drug re-exposure during abstinence, effects on cocaine-mediated behaviors, or potentially sexually dimorphic effects. Male and female rats were administered cocaine for 7 days and expression of sensitization was assessed by cocaine challenge after 21 days abstinence. On test days, 15 mg/kg i. p. cocaine was evaluated, whereas 30 mg/kg was administered on intervening days. Lower test doses avoid competition of non-motor behaviors with locomotion. In all measures where sensitization was expressed, ibudilast (7.5 and 10 mg/kg, i. p., b. i.d. for 3 days and once on test day) reversed this behavior to levels seen after acute exposure, but not below. There were some intriguing sexually dimorphic effects that were not a function of estrous cycle. Specifically, distance travelled in the center of the test arena and rearing only sensitized in male rats, and ibudilast reversed these behaviors to levels seen after acute cocaine exposure. In females, center distance travelled was reduced below acute cocaine levels by 7.5 mg/kg ibudilast. Increased distance travelled in the center versus periphery is thought to model anxiolytic-like behavior due to increased predation risk. Taken together, these data suggest that the clinical evaluation of ibudilast could be extended to cocaine use disorder.
Collapse
Affiliation(s)
- Ryan S. Poland
- Department of Psychiatry, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
| | - Yun Hahn
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
| | - Pamela E. Knapp
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
| | - Patrick M. Beardsley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
| | - M. Scott Bowers
- Department of Psychiatry, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
| |
Collapse
|
135
|
Crews FT, Vetreno RP, Broadwater MA, Robinson DL. Adolescent Alcohol Exposure Persistently Impacts Adult Neurobiology and Behavior. Pharmacol Rev 2016; 68:1074-1109. [PMID: 27677720 PMCID: PMC5050442 DOI: 10.1124/pr.115.012138] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Adolescence is a developmental period when physical and cognitive abilities are optimized, when social skills are consolidated, and when sexuality, adolescent behaviors, and frontal cortical functions mature to adult levels. Adolescents also have unique responses to alcohol compared with adults, being less sensitive to ethanol sedative-motor responses that most likely contribute to binge drinking and blackouts. Population studies find that an early age of drinking onset correlates with increased lifetime risks for the development of alcohol dependence, violence, and injuries. Brain synapses, myelination, and neural circuits mature in adolescence to adult levels in parallel with increased reflection on the consequence of actions and reduced impulsivity and thrill seeking. Alcohol binge drinking could alter human development, but variations in genetics, peer groups, family structure, early life experiences, and the emergence of psychopathology in humans confound studies. As adolescence is common to mammalian species, preclinical models of binge drinking provide insight into the direct impact of alcohol on adolescent development. This review relates human findings to basic science studies, particularly the preclinical studies of the Neurobiology of Adolescent Drinking in Adulthood (NADIA) Consortium. These studies focus on persistent adult changes in neurobiology and behavior following adolescent intermittent ethanol (AIE), a model of underage drinking. NADIA studies and others find that AIE results in the following: increases in adult alcohol drinking, disinhibition, and social anxiety; altered adult synapses, cognition, and sleep; reduced adult neurogenesis, cholinergic, and serotonergic neurons; and increased neuroimmune gene expression and epigenetic modifiers of gene expression. Many of these effects are specific to adolescents and not found in parallel adult studies. AIE can cause a persistence of adolescent-like synaptic physiology, behavior, and sensitivity to alcohol into adulthood. Together, these findings support the hypothesis that adolescent binge drinking leads to long-lasting changes in the adult brain that increase risks of adult psychopathology, particularly for alcohol dependence.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies (F.T.C., R.P.V., M.A.B., D.L.R.), Department of Psychiatry (F.T.C., D.L.R.), and Department of Pharmacology (F.T.C.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies (F.T.C., R.P.V., M.A.B., D.L.R.), Department of Psychiatry (F.T.C., D.L.R.), and Department of Pharmacology (F.T.C.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Margaret A Broadwater
- Bowles Center for Alcohol Studies (F.T.C., R.P.V., M.A.B., D.L.R.), Department of Psychiatry (F.T.C., D.L.R.), and Department of Pharmacology (F.T.C.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Donita L Robinson
- Bowles Center for Alcohol Studies (F.T.C., R.P.V., M.A.B., D.L.R.), Department of Psychiatry (F.T.C., D.L.R.), and Department of Pharmacology (F.T.C.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
136
|
Arredondo C, González M, Andrés ME, Gysling K. Opposite effects of acute and chronic amphetamine on Nurr1 and NF-κB p65 in the rat ventral tegmental area. Brain Res 2016; 1652:14-20. [PMID: 27687740 DOI: 10.1016/j.brainres.2016.09.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 09/20/2016] [Accepted: 09/21/2016] [Indexed: 12/21/2022]
Abstract
Dopamine neurons are overstimulated by drugs of abuse and suffer molecular alterations that lead to addiction behavior. Nurr1 is a transcription factor crucial for dopamine neurons survival and dopamine production, activating the transcription of key genes like tyrosine hydroxylase (TH). Interestingly, nuclear factor-kappa B (NF-κB) has emerged as a new Nurr1 partner in response to inflammatory stimulus. In this study we evaluated the effects of single and repeated amphetamine administration in the expression of Nurr1 and the NF-κB p65 subunit in the rat ventral tegmental area (VTA). We found that acute amphetamine treatment increased Nurr1, p65 and TH protein levels in the VTA. On the other hand, chronic amphetamine treatment decreased Nurr1 and p65 protein levels, but TH was unchanged. Mammalian reporter assays in cell lines showed that p65 represses Nurr1 transcriptional activity in an artificial promoter driven by Nurr1 response elements and in the native rat TH promoter. These results indicate that Nurr1 and NF-κB p65 factors are involved in the adaptive response of dopamine neurons to psychostimulants and that both transcription factors could be regulating Nurr1-dependent transactivation in the VTA.
Collapse
Affiliation(s)
- Cristian Arredondo
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - Marcela González
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - María Estela Andrés
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile.
| | - Katia Gysling
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile.
| |
Collapse
|
137
|
Cannon AR, Morris NL, Hammer AM, Curtis B, Remick DG, Yeligar SM, Poole L, Burnham EL, Wyatt TA, Molina PE, So-Armah K, Cisneros T, Wang G, Lang CH, Mandrekar P, Kovacs EJ, Choudhry MA. Alcohol and inflammatory responses: Highlights of the 2015 Alcohol and Immunology Research Interest Group (AIRIG) meeting. Alcohol 2016; 54:73-7. [PMID: 27522326 DOI: 10.1016/j.alcohol.2016.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 06/24/2016] [Indexed: 01/04/2023]
Abstract
On September 27, 2015 the 20th annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held as a satellite symposium at the annual meeting of the Society for Leukocyte Biology in Raleigh, NC. The 2015 meeting focused broadly on adverse effects of alcohol and alcohol-use disorders in multiple organ systems. Divided into two plenary sessions, AIRIG opened with the topic of pulmonary inflammation as a result of alcohol consumption, which was followed by alcohol's effect on multiple organs, including the brain and liver. With presentations showing the diverse range of underlying pathology and mechanisms associated with multiple organs as a result of alcohol consumption, AIRIG emphasized the importance of continued alcohol research, as its detrimental consequences are not limited to one or even two organs, but rather extend to the entire host as a whole.
Collapse
|
138
|
Suryanarayanan A, Carter JM, Landin JD, Morrow AL, Werner DF, Spigelman I. Role of interleukin-10 (IL-10) in regulation of GABAergic transmission and acute response to ethanol. Neuropharmacology 2016; 107:181-188. [PMID: 27016017 PMCID: PMC5076550 DOI: 10.1016/j.neuropharm.2016.03.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/08/2016] [Accepted: 03/21/2016] [Indexed: 12/27/2022]
Abstract
Mounting evidence indicates that ethanol (EtOH) exposure activates neuroimmune signaling. Alterations in pro-inflammatory cytokines after acute and chronic EtOH exposure have been heavily investigated. In contrast, little is known about the regulation of neurotransmission and/or modulation by anti-inflammatory cytokines in the brain after an acute EtOH exposure. Recent evidence suggests that interleukin-10 (IL-10), an anti-inflammatory cytokine, is upregulated during withdrawal from chronic EtOH exposure. In the present study, we show that IL-10 is increased early (1 h) after a single intoxicating dose of EtOH (5 g/kg, intragastric) in Sprague Dawley rats. We also show that IL-10 rapidly regulates GABAergic transmission in dentate gyrus neurons. In brain slice recordings, IL-10 application dose-dependently decreases miniature inhibitory postsynaptic current (mIPSC) area and frequency, and decreases the magnitude of the picrotoxin sensitive tonic current (Itonic), indicating both pre- and postsynaptic mechanisms. A PI3K inhibitor LY294002 (but not the negative control LY303511) ablated the inhibitory effects of IL-10 on mIPSC area and Itonic, but not on mIPSC frequency, indicating the involvement of PI3K in postsynaptic effects of IL-10 on GABAergic transmission. Lastly, we also identify a novel neurobehavioral regulation of EtOH sensitivity by IL-10, whereby IL-10 attenuates acute EtOH-induced hypnosis. These results suggest that EtOH causes an early release of IL-10 in the brain, which may contribute to neuronal hyperexcitability as well as disturbed sleep seen after binge exposure to EtOH. These results also identify IL-10 signaling as a potential therapeutic target in alcohol-use disorders and other CNS disorders where GABAergic transmission is altered.
Collapse
Affiliation(s)
- A Suryanarayanan
- Department of Pharmaceutical Sciences, University of the Sciences, Philadelphia College of Pharmacy, Philadelphia, PA 19104, USA.
| | - J M Carter
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY 13902, USA
| | - J D Landin
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY 13902, USA
| | - A L Morrow
- Departments of Psychiatry and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - D F Werner
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY 13902, USA
| | - I Spigelman
- Division of Oral Biology & Medicine, School of Dentistry, 63-078 CHS, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
139
|
Knapp DJ, Harper KM, Whitman BA, Zimomra Z, Breese GR. Stress and Withdrawal from Chronic Ethanol Induce Selective Changes in Neuroimmune mRNAs in Differing Brain Sites. Brain Sci 2016; 6:brainsci6030025. [PMID: 27472367 PMCID: PMC5039454 DOI: 10.3390/brainsci6030025] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/10/2016] [Accepted: 07/20/2016] [Indexed: 12/12/2022] Open
Abstract
Stress is a strong risk factor in alcoholic relapse and may exert effects that mimic aspects of chronic alcohol exposure on neurobiological systems. With the neuroimmune system becoming a prominent focus in the study of the neurobiological consequences of stress, as well as chronic alcohol exposure proving to be a valuable focus in this regard, the present study sought to compare the effects of stress and chronic ethanol exposure on induction of components of the neuroimmune system. Rats were exposed to either 1 h exposure to a mild stressor (restraint) or exposure to withdrawal from 15 days of chronic alcohol exposure (i.e., withdrawal from chronic ethanol, WCE) and assessed for neuroimmune mRNAs in brain. Restraint stress alone elevated chemokine (C–C motif) ligand 2 (CCL2), interleukin-1-beta (IL-1β), tumor necrosis factor alpha (TNFα) and toll-like receptor 4 (TLR4) mRNAs in the cerebral cortex within 4 h with a return to a control level by 24 h. These increases were not accompanied by an increase in corresponding proteins. Withdrawal from WCE also elevated cytokines, but did so to varying degrees across different cytokines and brain regions. In the cortex, stress and WCE induced CCL2, TNFα, IL-1β, and TLR4 mRNAs. In the hypothalamus, only WCE induced cytokines (CCL2 and IL-1β) while in the hippocampus, WCE strongly induced CCL2 while stress and WCE induced IL-1β. In the amygdala, only WCE induced CCL2. Finally—based on the previously demonstrated role of corticotropin-releasing factor 1 (CRF1) receptor inhibition in blocking WCE-induced cytokine mRNAs—the CRF1 receptor antagonist CP154,526 was administered to a subgroup of stressed rats and found to be inactive against induction of CCL2, TNFα, or IL-1β mRNAs. These differential results suggest that stress and WCE manifest broad neuroimmune effects in brain depending on the cytokine and brain region, and that CRF inhibition may not be a relevant mechanism in non-alcohol exposed animals. Overall, these effects are complex in terms of their neuroimmune targets and neuroanatomical specificity. Further investigation of the differential distribution of cytokine induction across neuroanatomical regions, individual cell types (e.g., neuronal phenotypes and glia), severity of chronic alcohol exposure, as well as across differing stress types may prove useful in understanding differential mechanisms of induction and for targeting select systems for pharmacotherapeutic intervention in alcoholism.
Collapse
Affiliation(s)
- Darin J Knapp
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Department of Psychiatry, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - Kathryn M Harper
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - Buddy A Whitman
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Curriculum in Neurobiology, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - Zachary Zimomra
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - George R Breese
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Department of Psychiatry, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Curriculum in Neurobiology, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Department of Pharmacology, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| |
Collapse
|
140
|
Merkel SF, Razmpour R, Lutton EM, Tallarida CS, Heldt NA, Cannella LA, Persidsky Y, Rawls SM, Ramirez SH. Adolescent Traumatic Brain Injury Induces Chronic Mesolimbic Neuroinflammation with Concurrent Enhancement in the Rewarding Effects of Cocaine in Mice during Adulthood. J Neurotrauma 2016; 34:165-181. [PMID: 27026056 DOI: 10.1089/neu.2015.4275] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Clinical psychiatric disorders of depression, anxiety, and substance abuse are most prevalent after traumatic brain injury (TBI). Pre-clinical research has focused on depression and anxiety post-injury; however, virtually no data exist examining whether the preference for illicit drugs is affected by traumatic injury in the developing adolescent brain. Using the controlled cortical impact (CCI) model of TBI and the conditioned place preference (CPP) assay, we tested the underlying hypothesis that brain injury during adolescence exacerbates the rewarding properties of cocaine in adulthood possibly through an active inflammatory status in the mesolimbic pathway. Six-week old, C57BL/6 mice sustained a single CCI-TBI to the right somatosensory cortex. CPP experiments with cocaine began 2 weeks post-TBI. Animals receiving cocaine displayed significant place preference shifts compared to saline controls. Further, within the cocaine-experienced cohort, moderate CCI-TBI during adolescence significantly increased the preference shift in adulthood when compared to naïve controls. Additionally, persistent neuroinflammatory responses were observed in the cortex, nucleus accumbens (NAc), and ventral tegmental area post-CCI-TBI. Significant increases in both astrocytic, glial fibrillary acidic protein, and microglial, ionization basic acid 1, markers were observed in the NAc at the end of CPP testing. Moreover, analysis using focused array gene expression panels identified the upregulation of numerous inflammatory genes in moderate CCI-TBI animals, compared to naïve controls, both in the cortex and NAc at 2 weeks post-TBI, before onset of cocaine administration. These results suggest that sustaining moderate TBI during adolescence may augment the rewarding effects of psychostimulants in adulthood, possibly by induction of chronic mesolimbic neuroinflammation.
Collapse
Affiliation(s)
- Steven F Merkel
- 1 Department of Pathology and Laboratory Medicine, Temple University School of Medicine , Philadelphia, Pennsylvania.,2 The Center for Substance Abuse Research, Temple University School of Medicine , Philadelphia, Pennsylvania
| | - Roshanak Razmpour
- 1 Department of Pathology and Laboratory Medicine, Temple University School of Medicine , Philadelphia, Pennsylvania
| | - Evan M Lutton
- 1 Department of Pathology and Laboratory Medicine, Temple University School of Medicine , Philadelphia, Pennsylvania
| | - Christopher S Tallarida
- 2 The Center for Substance Abuse Research, Temple University School of Medicine , Philadelphia, Pennsylvania.,4 Department of Pharmacology, Temple University School of Medicine , Philadelphia, Pennsylvania
| | - Nathan A Heldt
- 1 Department of Pathology and Laboratory Medicine, Temple University School of Medicine , Philadelphia, Pennsylvania
| | - Lee Anne Cannella
- 1 Department of Pathology and Laboratory Medicine, Temple University School of Medicine , Philadelphia, Pennsylvania
| | - Yuri Persidsky
- 1 Department of Pathology and Laboratory Medicine, Temple University School of Medicine , Philadelphia, Pennsylvania.,2 The Center for Substance Abuse Research, Temple University School of Medicine , Philadelphia, Pennsylvania
| | - Scott M Rawls
- 2 The Center for Substance Abuse Research, Temple University School of Medicine , Philadelphia, Pennsylvania.,4 Department of Pharmacology, Temple University School of Medicine , Philadelphia, Pennsylvania
| | - Servio H Ramirez
- 1 Department of Pathology and Laboratory Medicine, Temple University School of Medicine , Philadelphia, Pennsylvania.,2 The Center for Substance Abuse Research, Temple University School of Medicine , Philadelphia, Pennsylvania.,3 The Shriners Hospitals Pediatric Research Center, Temple University School of Medicine , Philadelphia, Pennsylvania
| |
Collapse
|
141
|
Marshall SA, Geil CR, Nixon K. Prior Binge Ethanol Exposure Potentiates the Microglial Response in a Model of Alcohol-Induced Neurodegeneration. Brain Sci 2016; 6:E16. [PMID: 27240410 PMCID: PMC4931493 DOI: 10.3390/brainsci6020016] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/12/2016] [Accepted: 05/16/2016] [Indexed: 12/20/2022] Open
Abstract
Excessive alcohol consumption results in neurodegeneration which some hypothesize is caused by neuroinflammation. One characteristic of neuroinflammation is microglial activation, but it is now well accepted that microglial activation may be pro- or anti-inflammatory. Recent work indicates that the Majchrowicz model of alcohol-induced neurodegeneration results in anti-inflammatory microglia, while intermittent exposure models with lower doses and blood alcohol levels produce microglia with a pro-inflammatory phenotype. To determine the effect of a repeated binge alcohol exposure, rats received two cycles of the four-day Majchrowicz model. One hemisphere was then used to assess microglia via immunohistochemistry and while the other was used for ELISAs of cytokines and growth factors. A single binge ethanol exposure resulted in low-level of microglial activation; however, a second binge potentiated the microglial response. Specifically, double binge rats had greater OX-42 immunoreactivity, increased ionized calcium-binding adapter molecule 1 (Iba-1+) cells, and upregulated tumor necrosis factor-α (TNF-α) compared with the single binge ethanol group. These data indicate that prior ethanol exposure potentiates a subsequent microglia response, which suggests that the initial exposure to alcohol primes microglia. In summary, repeated ethanol exposure, independent of other immune modulatory events, potentiates microglial activity.
Collapse
Affiliation(s)
- Simon Alex Marshall
- Department of Psychology & Neuroscience; University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Chelsea Rhea Geil
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA.
| | - Kimberly Nixon
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
142
|
Breese GR, Knapp DJ. Persistent adaptation by chronic alcohol is facilitated by neuroimmune activation linked to stress and CRF. Alcohol 2016; 52:9-23. [PMID: 27139233 PMCID: PMC4855305 DOI: 10.1016/j.alcohol.2016.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/10/2015] [Accepted: 01/24/2016] [Indexed: 01/11/2023]
Abstract
This review updates the conceptual basis for the association of alcohol abuse with an insidious adaptation that facilitates negative affect during withdrawal from chronic intermittent alcohol (CIA) exposure - a change that later supports sensitization of stress-induced anxiety following alcohol abstinence. The finding that a CRF1-receptor antagonist (CRF1RA) minimized CIA withdrawal-induced negative affect supported an association of alcohol withdrawal with a stress mechanism. The finding that repeated stresses or multiple CRF injections into selected brain sites prior to a single 5-day chronic alcohol (CA) exposure induced anxiety during withdrawal provided critical support for a linkage of CIA withdrawal with stress. The determination that CRF1RA injection into positive CRF-sensitive brain sites prevented CIA withdrawal-induced anxiety provided support that neural path integration maintains the persistent CIA adaptation. Based upon reports that stress increases neuroimmune function, an effort was undertaken to test whether cytokines would support the adaptation induced by stress/CA exposure. Twenty-four hours after withdrawal from CIA, cytokine mRNAs were found to be increased in cortex as well as other sites in brain. Further, repeated cytokine injections into previously identified brain sites substituted for stress and CRF induction of anxiety during CA withdrawal. Discovery that a CRF1RA prevented the brain cytokine mRNA increase induced by CA withdrawal provided critical evidence for CRF involvement in this neuroimmune induction after CA withdrawal. However, the CRF1RA did not block the stress increase in cytokine mRNA increases in controls. The latter data supported the hypothesis that distinct mechanisms linked to stress and CA withdrawal can support common neuroimmune functions within a brain site. As evidence evolves concerning neural involvement in brain neuroimmune function, a better understanding of the progressive adaptation associated with CIA exposure will advance new knowledge that could possibly lead to strategies to combat alcohol abuse.
Collapse
Affiliation(s)
- George R Breese
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Curriculum in Neurobiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; The UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA.
| | - Darin J Knapp
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA
| |
Collapse
|
143
|
Abstract
RATIONALE Alcoholism is a primary, chronic relapsing disease of brain reward, motivation, memory, and related circuitry. It is characterized by an individual's continued drinking despite negative consequences related to alcohol use, which is exemplified by alcohol use leading to clinically significant impairment or distress. Chronic alcohol consumption increases the expression of innate immune signaling molecules (ISMs) in the brain that alter cognitive processes and promote alcohol drinking. OBJECTIVES Unraveling the mechanisms of alcohol-induced neuroimmune gene induction is complicated by positive loops of multiple cytokines and other signaling molecules that converge on nuclear factor kappa-light-chain-enhancer of activated B cells and activator protein-1 leading to induction of additional neuroimmune signaling molecules that amplify and expand the expression of ISMs. RESULTS Studies from our laboratory employing reverse transcription polymerase chain reaction (RT-PCR) to assess mRNA, immunohistochemistry and Western blot analysis to assess protein expression, and others suggest that ethanol increases brain neuroimmune gene and protein expression through two distinct mechanisms involving (1) systemic induction of innate immune molecules that are transported from blood to the brain and (2) the direct release of high-mobility group box 1 (HMGB1) from neurons in the brain. Released HMGB1 signals through multiple receptors, particularly Toll-like receptor (TLR) 4, that potentiate cytokine receptor responses leading to a hyperexcitable state that disrupts neuronal networks and increases excitotoxic neuronal death. Innate immune gene activation in brain is persistent, consistent with the chronic relapsing disease that is alcoholism. Expression of HMGB1, TLRs, and other ISMs is increased several-fold in the human orbital frontal cortex, and expression of these molecules is highly correlated with each other as well as lifetime alcohol consumption and age of drinking onset. CONCLUSIONS The persistent and cumulative nature of alcohol on HMGB1 and TLR gene induction support their involvement in alcohol-induced long-term changes in brain function and neurodegeneration.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, CB# 7178, 1021 Thurston-Bowles Building, Chapel Hill, NC, 27599-7178, USA.
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, CB# 7178, 1021 Thurston-Bowles Building, Chapel Hill, NC, 27599-7178, USA
| |
Collapse
|
144
|
Weil ZM, Corrigan JD, Karelina K. Alcohol abuse after traumatic brain injury: Experimental and clinical evidence. Neurosci Biobehav Rev 2016; 62:89-99. [DOI: 10.1016/j.neubiorev.2016.01.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/16/2015] [Accepted: 01/21/2016] [Indexed: 01/06/2023]
|
145
|
Cadoni C. Fischer 344 and Lewis Rat Strains as a Model of Genetic Vulnerability to Drug Addiction. Front Neurosci 2016; 10:13. [PMID: 26903787 PMCID: PMC4746315 DOI: 10.3389/fnins.2016.00013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/11/2016] [Indexed: 01/02/2023] Open
Abstract
Today it is well acknowledged that both nature and nurture play important roles in the genesis of psychopathologies, including drug addiction. Increasing evidence suggests that genetic factors contribute for at least 40–60% of the variation in liability to drug dependence. Human genetic studies suggest that multiple genes of small effect, rather than single genes, contribute to the genesis of behavioral psychopathologies. Therefore, the use of inbred rat strains might provide a valuable tool to identify differences, linked to genotype, important in liability to addiction and related disorders. In this regard, Lewis and Fischer 344 inbred rats have been proposed as a model of genetic vulnerability to drug addiction, given their innate differences in sensitivity to the reinforcing and rewarding effects of drugs of abuse, as well their different responsiveness to stressful stimuli. This review will provide evidence in support of this model for the study of the genetic influence on addiction vulnerability, with particular emphasis on differences in mesolimbic dopamine (DA) transmission, rewarding and emotional function. It will be highlighted that Lewis and Fischer 344 rats differ not only in several indices of DA transmission and adaptive changes following repeated drug exposure, but also in hypothalamic-pituitary-adrenal (HPA) axis responsiveness, influencing not only the ability of the individual to cope with stressful events, but also interfering with rewarding and motivational processes, given the influence of corticosteroids on dopamine neuron functionality. Further differences between the two strains, as impulsivity or anxiousness, might contribute to their different proneness to addiction, and likely these features might be linked to their different DA neurotransmission plasticity. Although differences in other neurotransmitter systems might deserve further investigation, results from the reviewed studies might open new vistas in understanding aberrant deviations in reward and motivational functions.
Collapse
Affiliation(s)
- Cristina Cadoni
- Institute of Neuroscience, Cagliari Section, Department of Biomedical Sciences, National Research Council of ItalyCagliari, Italy; Centre of Excellence "Neurobiology of Dependence", University of CagliariCagliari, Italy
| |
Collapse
|
146
|
Marshall SA, Casachahua JD, Rinker JA, Blose AK, Lysle DT, Thiele TE. IL-1 receptor signaling in the basolateral amygdala modulates binge-like ethanol consumption in male C57BL/6J mice. Brain Behav Immun 2016; 51:258-267. [PMID: 26365025 PMCID: PMC4679505 DOI: 10.1016/j.bbi.2015.09.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/26/2015] [Accepted: 09/09/2015] [Indexed: 12/26/2022] Open
Abstract
Proinflammatory cytokines have been implicated in alcohol-induced neurodegeneration, but the role of the neuroimmune system in alcohol related behaviors has only recently come to the forefront. Herein, the effects of binge-like drinking on IL-1β mRNA and immunoreactivity within the amygdala were measured following the "drinking in the dark" (DID) paradigm, a model of binge-like ethanol drinking in C57BL/6J mice. Moreover, the role of IL-1 receptor signaling in the amygdala on ethanol consumption was assessed. Results indicated that a history of binge-like ethanol drinking promoted a significant increase of IL-1β mRNA expression within the amygdala, and immunohistochemistry analyses revealed that the basolateral amygdala (BLA), but not central amygdala (CeA), exhibited significantly increased IL-1β immunoreactivity. However, Fluoro-Jade® C labeling indicated that multiple cycles of the DID paradigm were not sufficient to elicit neuronal death. Bilateral infusions of IL-1 receptor antagonist (IL-1Ra) reduced ethanol consumption when infused into the BLA but not the CeA. These observations were specific to ethanol drinking as the IL-1Ra did not alter either sucrose drinking or open-field locomotor activity. The current findings highlight a specific role for IL-1 receptor signaling in modulating binge-like ethanol consumption and indicate that proinflammatory cytokines can be induced prior to dependence or any evidence of neuronal cell death. These findings provide a framework in which to understand how neuroimmune adaptations may alter ethanol consumption and therein contribute to alcohol abuse.
Collapse
Affiliation(s)
- S Alex Marshall
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - John D Casachahua
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jennifer A Rinker
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Allyson K Blose
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Donald T Lysle
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Todd E Thiele
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
147
|
García-Marchena N, Araos PF, Barrios V, Sánchez-Marín L, Chowen JA, Pedraz M, Castilla-Ortega E, Romero-Sanchiz P, Ponce G, Gavito AL, Decara J, Silva D, Torrens M, Argente J, Rubio G, Serrano A, de Fonseca FR, Pavón FJ. Plasma Chemokines in Patients with Alcohol Use Disorders: Association of CCL11 (Eotaxin-1) with Psychiatric Comorbidity. Front Psychiatry 2016; 7:214. [PMID: 28149283 PMCID: PMC5242327 DOI: 10.3389/fpsyt.2016.00214] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/26/2016] [Indexed: 12/16/2022] Open
Abstract
Recent studies have linked changes in peripheral chemokine concentrations to the presence of both addictive behaviors and psychiatric disorders. The present study further explore this link by analyzing the potential association of psychiatry comorbidity with alterations in the concentrations of circulating plasma chemokine in patients of both sexes diagnosed with alcohol use disorders (AUD). To this end, 85 abstinent subjects with AUD from an outpatient setting and 55 healthy subjects were evaluated for substance and mental disorders. Plasma samples were obtained to quantify chemokine concentrations [C-C motif (CC), C-X-C motif (CXC), and C-X3-C motif (CX3C) chemokines]. Abstinent AUD patients displayed a high prevalence of comorbid mental disorders (72%) and other substance use disorders (45%). Plasma concentrations of chemokines CXCL12/stromal cell-derived factor-1 (p < 0.001) and CX3CL1/fractalkine (p < 0.05) were lower in AUD patients compared to controls, whereas CCL11/eotaxin-1 concentrations were strongly decreased in female AUD patients (p < 0.001). In the alcohol group, CXCL8 concentrations were increased in patients with liver and pancreas diseases and there was a significant correlation to aspartate transaminase (r = +0.456, p < 0.001) and gamma-glutamyltransferase (r = +0.647, p < 0.001). Focusing on comorbid psychiatric disorders, we distinguish between patients with additional mental disorders (N = 61) and other substance use disorders (N = 38). Only CCL11 concentrations were found to be altered in AUD patients diagnosed with mental disorders (p < 0.01) with a strong main effect of sex. Thus, patients with mood disorders (N = 42) and/or anxiety (N = 16) had lower CCL11 concentrations than non-comorbid patients being more evident in women. The alcohol-induced alterations in circulating chemokines were also explored in preclinical models of alcohol use with male Wistar rats. Rats exposed to repeated ethanol (3 g/kg, gavage) had lower CXCL12 (p < 0.01) concentrations and higher CCL11 concentrations (p < 0.001) relative to vehicle-treated rats. Additionally, the increased CCL11 concentrations in rats exposed to ethanol were enhanced by the prior exposure to restraint stress (p < 0.01). Concordantly, acute ethanol exposure induced changes in CXCL12, CX3CL1, and CCL11 in the same direction to repeated exposure. These results clearly indicate a contribution of specific chemokines to the phenotype of AUD and a strong effect of sex, revealing a link of CCL11 to alcohol and anxiety/stress.
Collapse
Affiliation(s)
- Nuria García-Marchena
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Málaga, Spain; Facultad de Psicología, Universidad Complutense de Madrid, Madrid, Spain
| | - Pedro Fernando Araos
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga , Málaga , Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain; CIBER Fisiopatología de la obesidad y nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Sánchez-Marín
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga , Málaga , Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain; CIBER Fisiopatología de la obesidad y nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - María Pedraz
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga , Málaga , Spain
| | - Estela Castilla-Ortega
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga , Málaga , Spain
| | - Pablo Romero-Sanchiz
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga , Málaga , Spain
| | - Guillermo Ponce
- Servicio de Psiquiatría, Hospital Universitario 12 de Octubre , Madrid , Spain
| | - Ana L Gavito
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga , Málaga , Spain
| | - Juan Decara
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga , Málaga , Spain
| | - Daniel Silva
- Facultad de Psicología, Universidad Complutense de Madrid , Madrid , Spain
| | - Marta Torrens
- Institut de Neuropsiquiatria i Addiccions (INAD), Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain; Department of Psychiatry, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain; CIBER Fisiopatología de la obesidad y nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Gabriel Rubio
- Servicio de Psiquiatría, Hospital Universitario 12 de Octubre , Madrid , Spain
| | - Antonia Serrano
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga , Málaga , Spain
| | - Fernando Rodríguez de Fonseca
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Málaga, Spain; Facultad de Psicología, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisco Javier Pavón
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga , Málaga , Spain
| |
Collapse
|
148
|
Valenta JP, Gonzales RA. Chronic Intracerebroventricular Infusion of Monocyte Chemoattractant Protein-1 Leads to a Persistent Increase in Sweetened Ethanol Consumption During Operant Self-Administration But Does Not Influence Sucrose Consumption in Long-Evans Rats. Alcohol Clin Exp Res 2015; 40:187-95. [PMID: 26683974 DOI: 10.1111/acer.12928] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 10/12/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND Among the evidence implicating neuroimmune signaling in alcohol use disorders are increased levels of the chemokine monocyte chemoattractant protein-1 (MCP-1) in the brains of human alcoholics and animal models of alcohol abuse. However, it is not known whether neuroimmune signaling can directly increase ethanol (EtOH) consumption, and whether MCP-1 is involved in that mechanism. We designed experiments to determine whether MCP-1 signaling itself is sufficient to accelerate or increase EtOH consumption. Our hypothesis was that increasing MCP-1 signaling by directly infusing it into the brain would increase operant EtOH self-administration. METHODS We implanted osmotic minipumps to chronically infuse either one of several doses of MCP-1 or vehicle into the cerebral ventricles (intracerebroventricular) of Long-Evans rats and then tested them in the operant self-administration of a sweetened EtOH solution for 8 weeks. RESULTS There was a significant interaction between dose of MCP-1 and sweetened EtOH consumed across the first 4 weeks (while pumps were flowing) and across the 8-week experiment. Animals receiving the highest dose of MCP-1 (2 μg/d) were the highest consumers of EtOH during weeks 3 through 8. MCP-1 did not influence the acquisition of self-administration (measured across the first 5 days), the motivation to consume EtOH (time to lever press or progressive ratio), withdrawal-induced anxiety, or the consumption of sucrose alone. CONCLUSIONS We provide novel evidence that neuroimmune signaling can directly increase chronic operant EtOH self-administration, and that this increase persists beyond the administration of the cytokine. These data suggest that EtOH-induced increases in MCP-1, or increases in MCP-1 due to various other neuroimmune mechanisms, may further promote EtOH consumption. Continued research into this mechanism, particularly using models of alcohol dependence, will help determine whether targeting MCP-1 signaling has therapeutic potential in the treatment of alcohol use disorders.
Collapse
Affiliation(s)
- John P Valenta
- Division of Pharmacology/Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Rueben A Gonzales
- Division of Pharmacology/Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
149
|
Lemieux A, al'Absi M. Stress psychobiology in the context of addiction medicine: from drugs of abuse to behavioral addictions. PROGRESS IN BRAIN RESEARCH 2015; 223:43-62. [PMID: 26806770 DOI: 10.1016/bs.pbr.2015.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
In this chapter, we briefly review the basic biology of psychological stress and the stress response. We propose that psychological stress and the neurobiology of the stress response play in substance use initiation, maintenance, and relapse. The proposed mechanisms for this include, on the one hand, the complex interactions between biological mediators of the stress response and the dopaminergic reward system and, on the other hand, mediators of the stress response and other systems crucial in moderating key addiction-related behaviors such as endogenous opioids, the sympathetic-adrenal-medullary system, and endocannabinoids. Exciting new avenues of study including genomics, sex as a moderator of the stress response, and behavioral addictions (gambling, hypersexuality, dysfunctional internet use, and food as an addictive substance) are also briefly presented within the context of stress as a moderator of the addictive process.
Collapse
Affiliation(s)
| | - Mustafa al'Absi
- University of Minnesota School of Medicine, Duluth, MN, USA.
| |
Collapse
|
150
|
Lee KM, Chiu KB, Didier PJ, Baker KC, MacLean AG. Naltrexone treatment reverses astrocyte atrophy and immune dysfunction in self-harming macaques. Brain Behav Immun 2015; 50:288-297. [PMID: 26191654 PMCID: PMC4631668 DOI: 10.1016/j.bbi.2015.07.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/30/2015] [Accepted: 07/16/2015] [Indexed: 01/19/2023] Open
Abstract
The role of glia in the development and treatment of behavioral abnormalities is understudied. Recent reports have observed glial activation in several disorders, including depression, autism spectrum disorders and self-injurious behaviors (SIB). In the current study, we examined SIB in the physiologically and anatomically relevant nonhuman primate (NHP) model. At the Tulane National Primate Research Center (TNPRC), approximately 5% of singly housed macaques develop symptoms of SIB. We have previously demonstrated that naltrexone hydrochloride can be effective in reducing SIB. We have also demonstrated that the astrocytes of animals with SIB are distinctly atrophic and display heightened innate immune activation compared with control animals. We have added a third group of animals (five macaques identified with SIB and treated with oral naltrexone at a dose of 3.2mg/kg) to the previous cohort (six macaques with a history of SIB but not treated, and nine animals with no history of SIB) for this study. Gray and white matter astrocytes from frontal cortical tissue were examined following necropsy. Innate immune activation of astrocytes, which was increased in SIB animals, was markedly decreased in animals receiving naltrexone, as was atrophy of both grey and white matter astrocytes. This was concomitant with improved behavioral correlates. Preventing astrocyte activation in select areas of the brain to reduce injurious behavior is an innovative concept with implications for mental health studies. Differences in multiple areas of primate brain would help determine how self-injurious behavior develops. These studies suggest a stronger role for astrocytes in the cellular events associated with self-injurious behaviors.
Collapse
Affiliation(s)
- Kim M. Lee
- Tulane National Primate Research Center, Covington, LA, USA,Tulane Program in Biomedical Science, Tulane University School of Medicine, New Orleans, LA, USA
| | - Kevin B. Chiu
- Tulane National Primate Research Center, Covington, LA, USA,Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | | | - Kate C. Baker
- Tulane National Primate Research Center, Covington, LA, USA,Department of Psychology, Tulane University, New Orleans, LA, USA,Tulane Program in Neuroscience, Tulane University, New Orleans, LA, USA
| | - Andrew G. MacLean
- Tulane National Primate Research Center, Covington, LA, USA,Tulane Program in Biomedical Science, Tulane University School of Medicine, New Orleans, LA, USA,Tulane Program in Neuroscience, Tulane University, New Orleans, LA, USA,Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA,Corresponding author: Andrew G. MacLean, Tulane National Primate Research Center, Covington, LA, 70433. ‘phone: 985-871-6489
| |
Collapse
|