101
|
Glial and Neuroimmune Mechanisms as Critical Modulators of Drug Use and Abuse. Neuropsychopharmacology 2017; 42:156-177. [PMID: 27402494 PMCID: PMC5143481 DOI: 10.1038/npp.2016.121] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/24/2016] [Accepted: 06/28/2016] [Indexed: 12/26/2022]
Abstract
Drugs of abuse cause persistent alterations in synaptic plasticity that may underlie addiction behaviors. Evidence suggests glial cells have an essential and underappreciated role in the development and maintenance of drug abuse by influencing neuronal and synaptic functions in multifaceted ways. Microglia and astrocytes perform critical functions in synapse formation and refinement in the developing brain, and there is growing evidence that disruptions in glial function may be implicated in numerous neurological disorders throughout the lifespan. Linking evidence of function in health and under pathological conditions, this review will outline the glial and neuroimmune mechanisms that may contribute to drug-abuse liability, exploring evidence from opioids, alcohol, and psychostimulants. Drugs of abuse can activate microglia and astrocytes through signaling at innate immune receptors, which in turn influence neuronal function not only through secretion of soluble factors (eg, cytokines and chemokines) but also potentially through direct remodeling of the synapses. In sum, this review will argue that neural-glial interactions represent an important avenue for advancing our understanding of substance abuse disorders.
Collapse
|
102
|
Verburg-van Kemenade BML, Cohen N, Chadzinska M. Neuroendocrine-immune interaction: Evolutionarily conserved mechanisms that maintain allostasis in an ever-changing environment. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 66:2-23. [PMID: 27296493 DOI: 10.1016/j.dci.2016.05.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 05/02/2023]
Abstract
It has now become accepted that the immune system and neuroendocrine system form an integrated part of our physiology. Immunological defense mechanisms act in concert with physiological processes like growth and reproduction, energy intake and metabolism, as well as neuronal development. Not only are psychological and environmental stressors communicated to the immune system, but also, vice versa, the immune response and adaptation to a current pathogen challenge are communicated to the entire body, including the brain, to evoke adaptive responses (e.g., fever, sickness behavior) that ensure allocation of energy to fight the pathogen. This phenomenon is evolutionarily conserved. Hence it is both interesting and important to consider the evolutionary history of this bi-directional neuroendocrine-immune communication to reveal phylogenetically ancient or relatively recently acquired mechanisms. Indeed, such considerations have already disclosed an extensive "common vocabulary" of information pathways as well as molecules and their receptors used by both the neuroendocrine and immune systems. This review focuses on the principal mechanisms of bi-directional communication and the evidence for evolutionary conservation of the important physiological pathways involved.
Collapse
Affiliation(s)
- B M Lidy Verburg-van Kemenade
- Cell Biology and Immunology Group, Dept. of Animal Sciences, Wageningen University, P.O. Box 338, 6700 AH Wageningen, The Netherlands.
| | - Nicholas Cohen
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14620, USA
| | - Magdalena Chadzinska
- Department of Evolutionary Immunology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, PL30-387 Krakow, Poland
| |
Collapse
|
103
|
Harricharan R, Abboussi O, Daniels WM. Addiction: A dysregulation of satiety and inflammatory processes. PROGRESS IN BRAIN RESEARCH 2017; 235:65-91. [DOI: 10.1016/bs.pbr.2017.07.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
104
|
Reece AS, Norman A, Hulse GK. Cannabis exposure as an interactive cardiovascular risk factor and accelerant of organismal ageing: a longitudinal study. BMJ Open 2016; 6:e011891. [PMID: 27821595 PMCID: PMC5129004 DOI: 10.1136/bmjopen-2016-011891] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVES Many reports exist of the cardiovascular toxicity of smoked cannabis but none of arterial stiffness measures or vascular age (VA). In view of its diverse toxicology, the possibility that cannabis-exposed patients may be ageing more quickly requires investigation. DESIGN Cross-sectional and longitudinal, observational. Prospective. SETTING Single primary care addiction clinic in Brisbane, Australia. PARTICIPANTS 11 cannabis-only smokers, 504 tobacco-only smokers, 114 tobacco and cannabis smokers and 534 non-smokers. EXCLUSIONS known cardiovascular disease or therapy or acute exposure to alcohol, amphetamine, heroin or methadone. INTERVENTION Radial arterial pulse wave tonometry (AtCor, SphygmoCor, Sydney) performed opportunistically and sequentially on patients between 2006 and 2011. MAIN OUTCOME MEASURE Algorithmically calculated VA. SECONDARY OUTCOMES other central haemodynamic variables. RESULTS Differences between group chronological ages (CA, 30.47±0.48 to 40.36±2.44, mean±SEM) were controlled with linear regression. Between-group sex differences were controlled by single-sex analysis. Mean cannabis exposure among patients was 37.67±7.16 g-years. In regression models controlling for CA, Body Mass Index (BMI), time and inhalant group, the effect of cannabis use on VA was significant in males (p=0.0156) and females (p=0.0084). The effect size in males was 11.84%. A dose-response relationship was demonstrated with lifetime exposure (p<0.002) additional to that of tobacco and opioids. In both sexes, the effect of cannabis was robust to adjustment and was unrelated to its acute effects. Significant power interactions between cannabis exposure and the square and cube of CA were demonstrated (from p<0.002). CONCLUSIONS Cannabis is an interactive cardiovascular risk factor (additional to tobacco and opioids), shows a prominent dose-response effect and is robust to adjustment. Cannabis use is associated with an acceleration of the cardiovascular age, which is a powerful surrogate for the organismal-biological age. This likely underlies and bi-directionally interacts with its diverse toxicological profile and is of considerable public health and regulatory importance.
Collapse
Affiliation(s)
- Albert Stuart Reece
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Amanda Norman
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Gary Kenneth Hulse
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
105
|
Levandowski ML, Viola TW, Prado CH, Wieck A, Bauer ME, Brietzke E, Grassi-Oliveira R. Distinct behavioral and immunoendocrine parameters during crack cocaine abstinence in women reporting childhood abuse and neglect. Drug Alcohol Depend 2016; 167:140-8. [PMID: 27530287 DOI: 10.1016/j.drugalcdep.2016.08.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/02/2016] [Accepted: 08/04/2016] [Indexed: 01/01/2023]
Abstract
AIM To assess plasma levels of cortisol and cytokines between cocaine-dependent women with and without childhood maltreatment (CM) history during cocaine detoxification treatment. METHOD We assessed immunoendocrine and clinical parameters of 108 crack cocaine female users during 3 weeks of inpatient detoxification treatment, and 24 healthy women to obtain reference values. Women with (CM+, n=53) or without (CM-, n=55) CM history were identified answering the Childhood Trauma Questionnaire (CTQ). Blood samples and clinical assessment were collected before lunch during the first, second and third week post-treatment admission. Flow cytometry was used to assess TNF-α, IFN-γ, IL-2, IL-4, IL-6, IL-10, IL-17A plasma levels and ELISA assay was used to measure plasma cortisol levels. RESULTS At baseline, lower Th1 and Th17-related cytokines levels and higher Th2 cytokines levels were observed in crack cocaine users compared with reference values. Cytokines levels of cocaine dependents gradually became closer to reference values along detoxification treatment. However, when CM+ and CM- groups were compared, increased levels of IL-6, IL-4 and TNF-α across time were observed in CM+ group only. Additionally, a Th1/Th2 immune imbalance was observed within CM+ group, which was negatively correlated with the severity of the crack withdrawal. Finally, loading trauma exposure severity, immunoendocrine and clinical parameters in factor analysis, we identified three clusters of observed variables during detoxification: (1) systemic immunity and trauma exposure, (2) pro-inflammatory immunity and (3) behavior CONCLUSION Our results suggest the existence of an immunological phenotype variant associated with CM exposure during crack cocaine detoxification of women.
Collapse
Affiliation(s)
- Mateus Luz Levandowski
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), RS, Brazil; Post-Graduate Program in Psychology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (PUCRS), RS, Brazil
| | - Thiago Wendt Viola
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), RS, Brazil; Post-Graduate Program in Pediatrics and Children Health's, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (PUCRS), RS, Brazil
| | - Carine Hartmann Prado
- Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (PUCRS), RS, Brazil
| | - Andrea Wieck
- Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (PUCRS), RS, Brazil
| | - Moisés Evandro Bauer
- Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (PUCRS), RS, Brazil
| | - Elisa Brietzke
- Research Group in Behavioral Neuroscience of Bipolar Disorder, Departament of Psychiatry, Federal University of São Paulo Unifesp, São Paulo, SP, Brazil
| | - Rodrigo Grassi-Oliveira
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifical Catholic University of Rio Grande do Sul (PUCRS), RS, Brazil; Post-Graduate Program in Psychology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (PUCRS), RS, Brazil; Post-Graduate Program in Pediatrics and Children Health's, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (PUCRS), RS, Brazil.
| |
Collapse
|
106
|
Auvity S, Saba W, Goutal S, Leroy C, Buvat I, Cayla J, Caillé F, Bottlaender M, Cisternino S, Tournier N. Acute Morphine Exposure Increases the Brain Distribution of [18F]DPA-714, a PET Biomarker of Glial Activation in Nonhuman Primates. Int J Neuropsychopharmacol 2016; 20:67-71. [PMID: 27581167 PMCID: PMC5737475 DOI: 10.1093/ijnp/pyw077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 08/30/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The neuroinflammatory response to morphine exposure modulates its antinociceptive effects, tolerance, and dependence. Positron emission tomography radioligands for translocator protein-18kDa such as [18F]DPA-714 are noninvasive biomarkers of glial activation, a hallmark of neuroinflammation. METHODS [18F]DPA-714 positron emission tomography imaging was performed in 5 baboons at baseline and 2 hours after i.m. morphine injection (1 mg/kg). Brain kinetics and metabolite-corrected input function were measured to estimate [18F]DPA-714 brain distribution. RESULTS Morphine significantly increased [18F]DPA-714 brain distribution by a 1.3 factor (P<.05; paired t test). The effect was not restricted to opioid receptor-rich regions. Differences in baseline [18F]DPA-714 binding were observed among baboons. The response to morphine predominated in animals with the highest baseline uptake. CONCLUSIONS [18F]DPA-714 positron emission tomography imaging may be useful to noninvasively investigate the brain immune component of morphine pharmacology. Correlation between baseline brain distribution and subsequent response to morphine exposure suggest a role for priming parameters in controlling the neuroinflammatory properties of opioids.
Collapse
Affiliation(s)
- Sylvain Auvity
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France (Mr Auvity, Dr Saba, Mr Goutal, Dr Leroy, Dr Buvat, Mr Cayla, Dr Caillé, Dr Bottlaender, Dr Cisternino, and Dr Tournier); Variabilité de réponse aux psychotropes, Inserm, Université Paris Descartes, Faculté de pharmacie, Université Paris Diderot, UMR-S 1144, Paris,France (Mr Auvity and Dr Cisternino)
| | - Wadad Saba
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France (Mr Auvity, Dr Saba, Mr Goutal, Dr Leroy, Dr Buvat, Mr Cayla, Dr Caillé, Dr Bottlaender, Dr Cisternino, and Dr Tournier); Variabilité de réponse aux psychotropes, Inserm, Université Paris Descartes, Faculté de pharmacie, Université Paris Diderot, UMR-S 1144, Paris,France (Mr Auvity and Dr Cisternino)
| | - Sébastien Goutal
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France (Mr Auvity, Dr Saba, Mr Goutal, Dr Leroy, Dr Buvat, Mr Cayla, Dr Caillé, Dr Bottlaender, Dr Cisternino, and Dr Tournier); Variabilité de réponse aux psychotropes, Inserm, Université Paris Descartes, Faculté de pharmacie, Université Paris Diderot, UMR-S 1144, Paris,France (Mr Auvity and Dr Cisternino)
| | - Claire Leroy
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France (Mr Auvity, Dr Saba, Mr Goutal, Dr Leroy, Dr Buvat, Mr Cayla, Dr Caillé, Dr Bottlaender, Dr Cisternino, and Dr Tournier); Variabilité de réponse aux psychotropes, Inserm, Université Paris Descartes, Faculté de pharmacie, Université Paris Diderot, UMR-S 1144, Paris,France (Mr Auvity and Dr Cisternino)
| | - Irène Buvat
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France (Mr Auvity, Dr Saba, Mr Goutal, Dr Leroy, Dr Buvat, Mr Cayla, Dr Caillé, Dr Bottlaender, Dr Cisternino, and Dr Tournier); Variabilité de réponse aux psychotropes, Inserm, Université Paris Descartes, Faculté de pharmacie, Université Paris Diderot, UMR-S 1144, Paris,France (Mr Auvity and Dr Cisternino)
| | - Jérôme Cayla
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France (Mr Auvity, Dr Saba, Mr Goutal, Dr Leroy, Dr Buvat, Mr Cayla, Dr Caillé, Dr Bottlaender, Dr Cisternino, and Dr Tournier); Variabilité de réponse aux psychotropes, Inserm, Université Paris Descartes, Faculté de pharmacie, Université Paris Diderot, UMR-S 1144, Paris,France (Mr Auvity and Dr Cisternino)
| | - Fabien Caillé
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France (Mr Auvity, Dr Saba, Mr Goutal, Dr Leroy, Dr Buvat, Mr Cayla, Dr Caillé, Dr Bottlaender, Dr Cisternino, and Dr Tournier); Variabilité de réponse aux psychotropes, Inserm, Université Paris Descartes, Faculté de pharmacie, Université Paris Diderot, UMR-S 1144, Paris,France (Mr Auvity and Dr Cisternino)
| | - Michel Bottlaender
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France (Mr Auvity, Dr Saba, Mr Goutal, Dr Leroy, Dr Buvat, Mr Cayla, Dr Caillé, Dr Bottlaender, Dr Cisternino, and Dr Tournier); Variabilité de réponse aux psychotropes, Inserm, Université Paris Descartes, Faculté de pharmacie, Université Paris Diderot, UMR-S 1144, Paris,France (Mr Auvity and Dr Cisternino)
| | - Salvatore Cisternino
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France (Mr Auvity, Dr Saba, Mr Goutal, Dr Leroy, Dr Buvat, Mr Cayla, Dr Caillé, Dr Bottlaender, Dr Cisternino, and Dr Tournier); Variabilité de réponse aux psychotropes, Inserm, Université Paris Descartes, Faculté de pharmacie, Université Paris Diderot, UMR-S 1144, Paris,France (Mr Auvity and Dr Cisternino)
| | - Nicolas Tournier
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France (Mr Auvity, Dr Saba, Mr Goutal, Dr Leroy, Dr Buvat, Mr Cayla, Dr Caillé, Dr Bottlaender, Dr Cisternino, and Dr Tournier); Variabilité de réponse aux psychotropes, Inserm, Université Paris Descartes, Faculté de pharmacie, Université Paris Diderot, UMR-S 1144, Paris,France (Mr Auvity and Dr Cisternino)
| |
Collapse
|
107
|
Wang J, Zhao J, Liu Z, Guo F, Wang Y, Wang X, Zhang R, Vreugdenhil M, Lu C. Acute Ethanol Inhibition of γ Oscillations Is Mediated by Akt and GSK3β. Front Cell Neurosci 2016; 10:189. [PMID: 27582689 PMCID: PMC4987361 DOI: 10.3389/fncel.2016.00189] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 07/19/2016] [Indexed: 01/23/2023] Open
Abstract
Hippocampal network oscillations at gamma band frequency (γ, 30-80 Hz) are closely associated with higher brain functions such as learning and memory. Acute ethanol exposure at intoxicating concentrations (≥50 mM) impairs cognitive function. This study aimed to determine the effects and the mechanisms of acute ethanol exposure on γ oscillations in an in vitro model. Ethanol (25-100 mM) suppressed kainate-induced γ oscillations in CA3 area of the rat hippocampal slices, in a concentration-dependent, reversible manner. The ethanol-induced suppression was reduced by the D1R antagonist SCH23390 or the PKA inhibitor H89, was prevented by the Akt inhibitor triciribine or the GSk3β inhibitor SB415286, was enhanced by the NMDA receptor antagonist D-AP5, but was not affected by the MAPK inhibitor U0126 or PI3K inhibitor wortmanin. Our results indicate that the intracellular kinases Akt and GSk3β play a critical role in the ethanol-induced suppression of γ oscillations and reveal new cellular pathways involved in the ethanol-induced cognitive impairment.
Collapse
Affiliation(s)
- JianGang Wang
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical UniversityXinxiang, China; Department of Pathophysiology, Xinxiang Medical UniversityXinxiang, China
| | - JingXi Zhao
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical UniversityXinxiang, China; Psychiatric Hospital of Henan ProvinceXinxiang, China
| | - ZhiHua Liu
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical UniversityXinxiang, China; Psychiatric Hospital of Henan ProvinceXinxiang, China
| | - FangLi Guo
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical UniversityXinxiang, China; Department of Neurobiology and Physiology, Xinxiang Medical UniversityXinxiang, China
| | - Yali Wang
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical UniversityXinxiang, China; Department of Neurobiology and Physiology, Xinxiang Medical UniversityXinxiang, China
| | - Xiaofang Wang
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University Xinxiang, China
| | - RuiLing Zhang
- Psychiatric Hospital of Henan Province Xinxiang, China
| | - Martin Vreugdenhil
- Department of Psychology, Xinxiang Medical UniversityHenan, China; Department of Health Sciences, Birmingham City UniversityBirmingham, UK
| | - Chengbiao Lu
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical UniversityXinxiang, China; Psychiatric Hospital of Henan ProvinceXinxiang, China
| |
Collapse
|
108
|
Lebonville CL, Jones ME, Hutson LW, Cooper LB, Fuchs RA, Lysle DT. Acquisition of heroin conditioned immunosuppression requires IL-1 signaling in the dorsal hippocampus. Brain Behav Immun 2016; 56:325-34. [PMID: 27072068 PMCID: PMC4917416 DOI: 10.1016/j.bbi.2016.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/31/2016] [Accepted: 04/08/2016] [Indexed: 12/31/2022] Open
Abstract
Opioid users experience increased incidence of infection, which may be partially attributable to both direct opiate-immune interactions and conditioned immune responses. Previous studies have investigated the neural circuitry governing opioid conditioned immune responses, but work remains to elucidate the mechanisms mediating this effect. Our laboratory has previously shown that hippocampal IL-1 signaling, specifically, is required for the expression of heroin conditioned immunosuppression following learning. The current studies were designed to further characterize the role of hippocampal IL-1 in this phenomenon by manipulating IL-1 during learning. Experiment 1 tested whether hippocampal IL-1 is also required for the acquisition of heroin conditioned immunosuppression, while Experiment 2 tested whether hippocampal IL-1 is required for the expression of unconditioned heroin immunosuppression. We found that blocking IL-1 signaling in the dorsal hippocampus with IL-1RA during each conditioning session, but not on interspersed non-conditioning days, significantly attenuated the acquisition of heroin conditioned immunosuppression. Strikingly, we found that the same IL-1RA treatment did not alter unconditioned immunosuppression to a single dose of heroin. Thus, IL-1 signaling is not a critical component of the response to heroin but rather may play a role in the formation of the association between heroin and the context. Collectively, these studies suggest that IL-1 signaling, in addition to being involved in the expression of a heroin conditioned immune response, is also involved in the acquisition of this effect. Importantly, this effect is likely not due to blocking the response to the unconditioned stimulus since IL-1RA did not affect heroin's immunosuppressive effects.
Collapse
Affiliation(s)
- Christina L Lebonville
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, CB#3270, Chapel Hill, NC 27599-3270, USA
| | - Meghan E Jones
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, CB#3270, Chapel Hill, NC 27599-3270, USA
| | - Lee W Hutson
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, CB#3270, Chapel Hill, NC 27599-3270, USA
| | - Letty B Cooper
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, CB#3270, Chapel Hill, NC 27599-3270, USA
| | - Rita A Fuchs
- Washington State University College of Veterinary Medicine, Department of Integrative Physiology and Neuroscience, PO Box 647620, Pullman, WA 99164-7620, USA
| | - Donald T Lysle
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, CB#3270, Chapel Hill, NC 27599-3270, USA.
| |
Collapse
|
109
|
Nusslock R, Miller GE. Early-Life Adversity and Physical and Emotional Health Across the Lifespan: A Neuroimmune Network Hypothesis. Biol Psychiatry 2016; 80:23-32. [PMID: 26166230 PMCID: PMC4670279 DOI: 10.1016/j.biopsych.2015.05.017] [Citation(s) in RCA: 413] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 05/07/2015] [Accepted: 05/26/2015] [Indexed: 12/29/2022]
Abstract
Children who experience chronic stressors are vulnerable to emotional and physical health problems across the lifespan. This phenomenon raises questions for scientists and clinicians alike. How does adversity get under the skin of the developing child? Through what mechanisms does it confer vulnerability to a heterogeneous set of mental and physical illnesses? And how does it instantiate risk across different life stages, engendering vulnerability to conditions that develop shortly after stressor exposure-like depression-and conditions that manifest decades later, like heart disease? Although answers to these questions have started to emerge, research has typically focused on single diseases or organ systems. To understand the plethora of health problems associated with childhood adversity, we argue that the field needs a second generation of research that recognizes multidirectional transactions among biological systems. To help facilitate this process, we propose a neuroimmune network hypothesis as a heuristic framework for organizing knowledge from disparate literatures and as a springboard for generating integrative research. Drawing on existing data, we argue that early-life adversity amplifies crosstalk between peripheral inflammation and neural circuitries subserving threat-related, reward-related, and executive control-related processes. This crosstalk results in chronic low-grade inflammation, thereby contributing to adiposity, insulin resistance, and other predisease states. In the brain, inflammatory mediators act on cortico-amygdala threat and cortico-basal ganglia reward, circuitries in a manner that predisposes individuals to self-medicating behaviors like smoking, drug use, and consumption of high-fat diets. Acting in concert with inflammation, these behaviors accelerate the pathogenesis of emotional and physical health problems.
Collapse
Affiliation(s)
| | - Gregory E. Miller
- Department of Psychology, Northwestern University,Institute for Policy Research, Northwestern University
| |
Collapse
|
110
|
Worley MJ, Swanson AN, Heinzerling KG, Roche DJO, Shoptaw S. Ibudilast attenuates subjective effects of methamphetamine in a placebo-controlled inpatient study. Drug Alcohol Depend 2016; 162:245-50. [PMID: 26993372 PMCID: PMC5349508 DOI: 10.1016/j.drugalcdep.2016.02.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 02/22/2016] [Accepted: 02/24/2016] [Indexed: 10/22/2022]
Abstract
BACKGROUND Despite numerous clinical trials no efficacious medications for methamphetamine (MA) have been identified. Neuroinflammation, which has a role in MA-related reward and neurodegeneration, is a novel MA pharmacotherapy target. Ibudilast inhibits activation of microglia and pro-inflammatory cytokines and has reduced MA self-administration in preclinical research. This study examined whether ibudilast would reduce subjective effects of MA in humans. METHODS Adult, non-treatment seeking, MA-dependent volunteers (N=11) received oral placebo, moderate ibudilast (40 mg), and high-dose ibudilast (100mg) via twice-daily dosing for 7 days each in an inpatient setting. Following infusions of saline, MA 15 mg, and MA 30 mg participants rated 12 subjective drug effects on a visual analog scale (VAS). RESULTS As demonstrated by statistically-significant ibudilast × MA condition interactions (p<.05), ibudilast reduced several MA-related subjective effects including High, Effect (i.e., any drug effect), Good, Stimulated and Like. The ibudilast-related reductions were most pronounced in the MA 30 mg infusions, with ibudilast 100mg significantly reducing Effect (97.5% CI [-12.54, -2.27]), High (97.5% CI [-12.01, -1.65]), and Good (97.5% CI [-11.20, -0.21]), compared to placebo. CONCLUSIONS Ibudilast appeared to reduce reward-related subjective effects of MA in this early-stage study, possibly due to altering the processes of neuroinflammation involved in MA reward. Given this novel mechanism of action and the absence of an efficacious medication for MA dependence, ibudilast warrants further study to evaluate its clinical efficacy.
Collapse
Affiliation(s)
- Matthew J Worley
- Department of Family Medicine, University of California, Los Angeles, 10880 Wilshire Blvd., Suite 1800, Los Angeles, CA 90024, United States.
| | | | - Keith G Heinzerling
- Department of Family Medicine, University of California, Los Angeles, 10880 Wilshire Blvd., Suite 1800, Los Angeles, CA 90024, United States
| | - Daniel J O Roche
- Department of Psychology, University of California, Los Angeles, Franz Hall, Los Angeles, CA 90095, United States
| | - Steve Shoptaw
- Department of Family Medicine, University of California, Los Angeles, 10880 Wilshire Blvd., Suite 1800, Los Angeles, CA 90024, United States
| |
Collapse
|
111
|
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: 4.2] [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]
|
112
|
Wang X, Zhang Y, Peng Y, Hutchinson MR, Rice KC, Yin H, Watkins LR. Pharmacological characterization of the opioid inactive isomers (+)-naltrexone and (+)-naloxone as antagonists of toll-like receptor 4. Br J Pharmacol 2016; 173:856-69. [PMID: 26603732 DOI: 10.1111/bph.13394] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 11/11/2015] [Accepted: 11/11/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The toll-like receptor TLR4 is involved in neuropathic pain and in drug reward and reinforcement. The opioid inactive isomers (+)-naltrexone and (+)-naloxone act as TLR4 antagonists, reversing neuropathic pain and reducing opioid and cocaine reward and reinforcement. However, how these agents modulate TLR4 signalling is not clear. Here, we have elucidated the molecular mechanism of (+)-naltrexone and (+)-naloxone on TLR4 signalling. EXPERIMENTAL APPROACH BV-2 mouse microglial cell line, primary rat microglia and primary rat peritoneal macrophages were treated with LPS and TLR4 signalling inhibitors. Effects were measured using Western blotting, luciferase reporter assays, fluorescence microscopy and ELISA KEY RESULTS: (+)-Naltrexone and (+)-naloxone were equi-potent inhibitors of the LPS-induced TLR4 downstream signalling and induction of the pro-inflammatory factors NO and TNF-α. Similarly, (+)-naltrexone or (+)-naloxone inhibited production of reactive oxygen species and increased microglial phagocytosis, induced by LPS. However, (+)-naltrexone and (+)-naloxone did not directly inhibit the increased production of IL-1β, induced by LPS. The drug interaction of (+)-naloxone and (+)-naltrexone was additive. (+)-Naltrexone or (+)-naloxone inhibited LPS-induced activation of IFN regulatory factor 3 and production of IFN-β. However, they did not inhibit TLR4 signalling via the activation of either NF-κB, p38 or JNK in these cellular models. CONCLUSIONS AND IMPLICATIONS (+)-Naltrexone and (+)-naloxone were TRIF-IFN regulatory factor 3 axis-biased TLR4 antagonists. They blocked TLR4 downstream signalling leading to NO, TNF-α and reactive oxygen species. This pattern may explain, at least in part, the in vivo therapeutic effects of (+)-naltrexone and (+)-naloxone.
Collapse
Affiliation(s)
- X Wang
- Chemical Biology Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Y Zhang
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - Y Peng
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - M R Hutchinson
- Discipline of Physiology, School of Medical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - K C Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20892, USA
| | - H Yin
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - L R Watkins
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, 80309, USA
| |
Collapse
|
113
|
Jacobsen JHW, Hutchinson MR, Mustafa S. Drug addiction: targeting dynamic neuroimmune receptor interactions as a potential therapeutic strategy. Curr Opin Pharmacol 2016; 26:131-7. [DOI: 10.1016/j.coph.2015.10.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/28/2015] [Accepted: 10/30/2015] [Indexed: 01/29/2023]
|
114
|
Abstract
UNLABELLED Treating pain is one of the most difficult challenges in medicine and a key facet of disease management. The isolation of morphine by Friedrich Sertürner in 1804 added an essential pharmacological tool in the treatment of pain and spawned the discovery of a new class of drugs known collectively as opioid analgesics. Revered for their potent pain-relieving effects, even Morpheus the god of dreams could not have dreamt that his opium tincture would be both a gift and a burden to humankind. To date, morphine and other opioids remain essential analgesics for alleviating pain. However, their use is plagued by major side effects, such as analgesic tolerance (diminished pain-relieving effects), hyperalgesia (increased pain sensitivity), and drug dependence. This review highlights recent advances in understanding the key causes of these adverse effects and explores the effect of chronic pain on opioid reward. SIGNIFICANCE STATEMENT Chronic pain is pervasive and afflicts >100 million Americans. Treating pain in these individuals is notoriously difficult and often requires opioids, one of the most powerful and effective classes of drugs used for controlling pain. However, their use is plagued by major side effects, such as a loss of pain-relieving effects (analgesic tolerance), paradoxical pain (hyperalgesia), and addiction. Despite the potential side effects, opioids remain the pharmacological cornerstone of modern pain therapy. This review highlights recent breakthroughs in understanding the key causes of these adverse effects and explores the cellular control of opioid systems in reward and aversion. The findings will challenge traditional views of the good, the bad, and the ugly of opioids.
Collapse
|
115
|
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: 6.0] [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
|
116
|
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: 2.0] [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
|
117
|
Huckans M, Fuller BE, Chalker ALN, Adams M, Loftis JM. Plasma Inflammatory Factors Are Associated with Anxiety, Depression, and Cognitive Problems in Adults with and without Methamphetamine Dependence: An Exploratory Protein Array Study. Front Psychiatry 2015; 6:178. [PMID: 26732994 PMCID: PMC4683192 DOI: 10.3389/fpsyt.2015.00178] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/04/2015] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES It is hypothesized that immune factors influence addictive behaviors and contribute to relapse. The primary study objectives were to (1) compare neuropsychiatric symptoms across adults with active methamphetamine (MA) dependence, in early remission from MA dependence, and with no history of substance dependence, (2) determine whether active or recent MA dependence affects the expression of immune factors, and (3) evaluate the association between immune factor levels and neuropsychiatric symptoms. METHODS A cross-sectional study was conducted using between group comparisons and regression analyses to investigate associations among variables. Eighty-four adults were recruited into control (CTL) (n = 31), MA-active (n = 17), or MA-remission (n = 36) groups. Participants completed self-report measures of anxiety, depression, and memory complaints and objective tests of attention and executive function. Blood samples were collected, and a panel of immune factors was measured using multiplex technology. RESULTS Relative to CTLs, MA-dependent adults evidenced greater anxiety and depression during active use (p < 0.001) and remission (p < 0.007), and more attention, memory, and executive problems during remission (p < 0.01) but not active dependence. Regression analyses identified 10 immune factors (putatively associated with cytokine-cytokine receptor interactions) associated with anxiety, depression, and memory problems. CONCLUSION While psychiatric symptoms are present during active MA dependence and remission, at least some cognitive difficulties emerge only during remission. Altered expression of a network of immune factors contributes to neuropsychiatric symptom severity.
Collapse
Affiliation(s)
- Marilyn Huckans
- Research and Development Service, VA Portland Health Care System, Portland, OR, USA
- Mental Health and Clinical Neurosciences Division, VA Portland Health Care System, Portland, OR, USA
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Bret E. Fuller
- Research and Development Service, VA Portland Health Care System, Portland, OR, USA
- Mental Health and Clinical Neurosciences Division, VA Portland Health Care System, Portland, OR, USA
| | - Alison L. N. Chalker
- Research and Development Service, VA Portland Health Care System, Portland, OR, USA
| | - Madeleine Adams
- Research and Development Service, VA Portland Health Care System, Portland, OR, USA
| | - Jennifer M. Loftis
- Research and Development Service, VA Portland Health Care System, Portland, OR, USA
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
118
|
DAT isn't all that: cocaine reward and reinforcement require Toll-like receptor 4 signaling. Mol Psychiatry 2015; 20:1525-37. [PMID: 25644383 PMCID: PMC4523496 DOI: 10.1038/mp.2014.177] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/16/2014] [Accepted: 11/12/2014] [Indexed: 01/13/2023]
Abstract
The initial reinforcing properties of drugs of abuse, such as cocaine, are largely attributed to their ability to activate the mesolimbic dopamine system. Resulting increases in extracellular dopamine in the nucleus accumbens (NAc) are traditionally thought to result from cocaine's ability to block dopamine transporters (DATs). Here we demonstrate that cocaine also interacts with the immunosurveillance receptor complex, Toll-like receptor 4 (TLR4), on microglial cells to initiate central innate immune signaling. Disruption of cocaine signaling at TLR4 suppresses cocaine-induced extracellular dopamine in the NAc, as well as cocaine conditioned place preference and cocaine self-administration. These results provide a novel understanding of the neurobiological mechanisms underlying cocaine reward/reinforcement that includes a critical role for central immune signaling, and offer a new target for medication development for cocaine abuse treatment.
Collapse
|
119
|
Mediouni S, Marcondes MCG, Miller C, McLaughlin JP, Valente ST. The cross-talk of HIV-1 Tat and methamphetamine in HIV-associated neurocognitive disorders. Front Microbiol 2015; 6:1164. [PMID: 26557111 PMCID: PMC4615951 DOI: 10.3389/fmicb.2015.01164] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/07/2015] [Indexed: 12/15/2022] Open
Abstract
Antiretroviral therapy has dramatically improved the lives of human immunodeficiency virus 1 (HIV-1) infected individuals. Nonetheless, HIV-associated neurocognitive disorders (HAND), which range from undetectable neurocognitive impairments to severe dementia, still affect approximately 50% of the infected population, hampering their quality of life. The persistence of HAND is promoted by several factors, including longer life expectancies, the residual levels of virus in the central nervous system (CNS) and the continued presence of HIV-1 regulatory proteins such as the transactivator of transcription (Tat) in the brain. Tat is a secreted viral protein that crosses the blood–brain barrier into the CNS, where it has the ability to directly act on neurons and non-neuronal cells alike. These actions result in the release of soluble factors involved in inflammation, oxidative stress and excitotoxicity, ultimately resulting in neuronal damage. The percentage of methamphetamine (MA) abusers is high among the HIV-1-positive population compared to the general population. On the other hand, MA abuse is correlated with increased viral replication, enhanced Tat-mediated neurotoxicity and neurocognitive impairments. Although several strategies have been investigated to reduce HAND and MA use, no clinically approved treatment is currently available. Here, we review the latest findings of the effects of Tat and MA in HAND and discuss a few promising potential therapeutic developments.
Collapse
Affiliation(s)
- Sonia Mediouni
- Department of Infectious Diseases, The Scripps Research Institute , Jupiter, FL, USA
| | | | - Courtney Miller
- Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, FL, USA ; Department of Neuroscience, The Scripps Research Institute , Jupiter, FL, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, University of Florida , Gainesville, FL, USA
| | - Susana T Valente
- Department of Infectious Diseases, The Scripps Research Institute , Jupiter, FL, USA
| |
Collapse
|
120
|
Dash S, Balasubramaniam M, Villalta F, Dash C, Pandhare J. Impact of cocaine abuse on HIV pathogenesis. Front Microbiol 2015; 6:1111. [PMID: 26539167 PMCID: PMC4611962 DOI: 10.3389/fmicb.2015.01111] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 09/25/2015] [Indexed: 12/13/2022] Open
Abstract
Over 1.2 million people in the United States are infected with the human immunodeficiency virus type 1 (HIV-1). Tremendous progress has been made over the past three decades on many fronts in the prevention and treatment of HIV-1 disease. However, HIV-1 infection is incurable and antiretroviral drugs continue to remain the only effective treatment option for HIV infected patients. Unfortunately, only three out of ten HIV-1 infected individuals in the US have the virus under control. Thus, majority of HIV-1 infected individuals in the US are either unaware of their infection status or not connected/retained to care or are non-adherent to antiretroviral therapy (ART). This national public health crisis, as well as the ongoing global HIV/AIDS pandemic, is further exacerbated by substance abuse, which serves as a powerful cofactor at every stage of HIV/AIDS including transmission, diagnosis, pathogenesis, and treatment. Clinical studies indicate that substance abuse may increase viral load, accelerate disease progression and worsen AIDS-related mortality even among ART-adherent patients. However, confirming a direct causal link between substance abuse and HIV/AIDS in human patients remains a highly challenging endeavor. In this review we will discuss the recent and past developments in clinical and basic science research on the effects of cocaine abuse on HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Sabyasachi Dash
- Center for AIDS Health Disparities Research, Meharry Medical College , Nashville, TN, USA ; School of Graduate Studies and Research, Meharry Medical College , Nashville, TN, USA
| | - Muthukumar Balasubramaniam
- Center for AIDS Health Disparities Research, Meharry Medical College , Nashville, TN, USA ; Department of Biochemistry and Cancer Biology, Meharry Medical College , Nashville, TN, USA
| | - Fernando Villalta
- Center for AIDS Health Disparities Research, Meharry Medical College , Nashville, TN, USA ; School of Graduate Studies and Research, Meharry Medical College , Nashville, TN, USA ; Department of Microbiology and Immunology, Meharry Medical College , Nashville, TN, USA
| | - Chandravanu Dash
- Center for AIDS Health Disparities Research, Meharry Medical College , Nashville, TN, USA ; School of Graduate Studies and Research, Meharry Medical College , Nashville, TN, USA ; Department of Biochemistry and Cancer Biology, Meharry Medical College , Nashville, TN, USA
| | - Jui Pandhare
- Center for AIDS Health Disparities Research, Meharry Medical College , Nashville, TN, USA ; School of Graduate Studies and Research, Meharry Medical College , Nashville, TN, USA ; Department of Microbiology and Immunology, Meharry Medical College , Nashville, TN, USA
| |
Collapse
|
121
|
Karoly HC, YorkWilliams SL, Hutchison KE. Clinical Neuroscience of Addiction: Similarities and Differences Between Alcohol and Other Drugs. Alcohol Clin Exp Res 2015; 39:2073-84. [DOI: 10.1111/acer.12884] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/21/2015] [Indexed: 01/14/2023]
Affiliation(s)
- Hollis C. Karoly
- Department of Psychology and Neuroscience University of Colorado, Boulder Boulder Colorado
| | - Sophie L. YorkWilliams
- Department of Psychology and Neuroscience University of Colorado, Boulder Boulder Colorado
| | - Kent E. Hutchison
- Department of Psychology and Neuroscience University of Colorado, Boulder Boulder Colorado
| |
Collapse
|
122
|
Araos P, Pedraz M, Serrano A, Lucena M, Barrios V, García-Marchena N, Campos-Cloute R, Ruiz JJ, Romero P, Suárez J, Baixeras E, de la Torre R, Montesinos J, Guerri C, Rodríguez-Arias M, Miñarro J, Martínez-Riera R, Torrens M, Chowen JA, Argente J, Mason BJ, Pavón FJ, Rodríguez de Fonseca F. Plasma profile of pro-inflammatory cytokines and chemokines in cocaine users under outpatient treatment: influence of cocaine symptom severity and psychiatric co-morbidity. Addict Biol 2015; 20:756-72. [PMID: 24854157 DOI: 10.1111/adb.12156] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The treatment for cocaine use constitutes a clinical challenge because of the lack of appropriate therapies and the high rate of relapse. Recent evidence indicates that the immune system might be involved in the pathogenesis of cocaine addiction and its co-morbid psychiatric disorders. This work examined the plasma pro-inflammatory cytokine and chemokine profile in abstinent cocaine users (n = 82) who sought outpatient cocaine treatment and age/sex/body mass-matched controls (n = 65). Participants were assessed with the diagnostic interview Psychiatric Research Interview for Substance and Mental Diseases according to the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision (DSM-IV-TR). Tumor necrosis factor-alpha, chemokine (C-C motif) ligand 2/monocyte chemotactic protein-1 and chemokine (C-X-C motif) ligand 12 (CXCL12)/stromal cell-derived factor-1 (SDF-1) were decreased in cocaine users, although all cytokines were identified as predictors of a lifetime pathological use of cocaine. Interleukin-1 beta (IL-1β), chemokine (C-X3-C motif) ligand 1 (CX3CL1)/fractalkine and CXCL12/SDF-1 positively correlated with the cocaine symptom severity when using the DSM-IV-TR criteria for cocaine abuse/dependence. These cytokines allowed the categorization of the outpatients into subgroups according to severity, identifying a subgroup of severe cocaine users (9-11 criteria) with increased prevalence of co-morbid psychiatric disorders [mood (54%), anxiety (32%), psychotic (30%) and personality (60%) disorders]. IL-1β was observed to be increased in users with such psychiatric disorders relative to those users with no diagnosis. In addition to these clinical data, studies in mice demonstrated that plasma IL-1β, CX3CL1 and CXCL12 were also affected after acute and chronic cocaine administration, providing a preclinical model for further research. In conclusion, cocaine exposure modifies the circulating levels of pro-inflammatory mediators. Plasma cytokine/chemokine monitoring could improve the stratification of cocaine consumers seeking treatment and thus facilitate the application of appropriate interventions, including management of heightened risk of psychiatric co-morbidity. Further research is necessary to elucidate the role of the immune system in the etiology of cocaine addiction.
Collapse
Affiliation(s)
- Pedro Araos
- Unidad Gestión Clínica de Salud Mental; Instituto de Investigación Biomédica de Málaga (IBIMA); Hospital Regional Universitario de Málaga; Málaga Spain
| | - María Pedraz
- Unidad Gestión Clínica de Salud Mental; Instituto de Investigación Biomédica de Málaga (IBIMA); Hospital Regional Universitario de Málaga; Málaga Spain
| | - Antonia Serrano
- Unidad Gestión Clínica de Salud Mental; Instituto de Investigación Biomédica de Málaga (IBIMA); Hospital Regional Universitario de Málaga; Málaga Spain
| | - Miguel Lucena
- 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
| | - Nuria García-Marchena
- Unidad Gestión Clínica de Salud Mental; Instituto de Investigación Biomédica de Málaga (IBIMA); Hospital Regional Universitario de Málaga; Málaga Spain
| | | | | | - Pablo Romero
- Unidad Gestión Clínica de Salud Mental; Instituto de Investigación Biomédica de Málaga (IBIMA); Hospital Regional Universitario de Málaga; Málaga Spain
| | - Juan Suárez
- Unidad Gestión Clínica de Salud Mental; Instituto de Investigación Biomédica de Málaga (IBIMA); Hospital Regional Universitario de Málaga; Málaga Spain
| | - Elena Baixeras
- Unidad Gestión Clínica de Salud Mental; Instituto de Investigación Biomédica de Málaga (IBIMA); Hospital Regional Universitario de Málaga; Málaga Spain
| | - Rafael de la Torre
- Neurosciences Program; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM); Barcelona Spain
- Facultat de Ciencies de la Salut i de la Vida; Universitat Pompeu Fabra (CEXS-UPF); Barcelona Spain
| | - Jorge Montesinos
- Department of Cellular Pathology; Príncipe Felipe Research Centre; Valencia Spain
| | - Consuelo Guerri
- Department of Cellular Pathology; Príncipe Felipe Research Centre; Valencia Spain
| | - Marta Rodríguez-Arias
- Unidad de Investigación Psicobiología de las Drogodependencias; Facultad de Psicología; Universitat de Valencia; Valencia Spain
| | - José Miñarro
- Unidad de Investigación Psicobiología de las Drogodependencias; Facultad de Psicología; Universitat de Valencia; Valencia Spain
| | - Roser Martínez-Riera
- Neurosciences Program; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM); Barcelona Spain
- Institut de Neuropsiquiatria i Addiccions (INAD) del Parc de Salut MAR; Barcelona Spain
- Department of Psychiatry; Universitat Autònoma de Barcelona (UAB); Barcelona Spain
| | - Marta Torrens
- Neurosciences Program; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM); Barcelona Spain
- Institut de Neuropsiquiatria i Addiccions (INAD) del Parc de Salut MAR; Barcelona Spain
- Department of Psychiatry; Universitat Autònoma de Barcelona (UAB); Barcelona Spain
| | - Julie A. Chowen
- Department of Endocrinology; Hospital Infantil Universitario Niño Jesús; Madrid Spain
| | - Jesús Argente
- Department of Endocrinology; Hospital Infantil Universitario Niño Jesús; Madrid Spain
| | - Barbara J. Mason
- Committee on the Neurobiology of Addictive Disorders; The Scripps Research Institute (TSRI); La Jolla CA USA
| | - Francisco J. Pavón
- 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
| |
Collapse
|
123
|
Selfridge BR, Wang X, Zhang Y, Yin H, Grace PM, Watkins LR, Jacobson AE, Rice KC. Structure-Activity Relationships of (+)-Naltrexone-Inspired Toll-like Receptor 4 (TLR4) Antagonists. J Med Chem 2015; 58:5038-52. [PMID: 26010811 DOI: 10.1021/acs.jmedchem.5b00426] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Activation of Toll-like receptors has been linked to neuropathic pain and opioid dependence. (+)-Naltrexone acts as a Toll-like receptor 4 (TLR4) antagonist and has been shown to reverse neuropathic pain in rat studies. We designed and synthesized compounds based on (+)-naltrexone and (+)-noroxymorphone and evaluated their TLR4 antagonist activities by their effects on inhibiting lipopolysaccharide (LPS) induced TLR4 downstream nitric oxide (NO) production in microglia BV-2 cells. Alteration of the N-substituent in (+)-noroxymorphone gave us a potent TLR4 antagonist. The most promising analog, (+)-N-phenethylnoroxymorphone ((4S,4aR,7aS,12bR)-4a,9-dihydroxy-3-phenethyl-2,3,4,4a,5,6-hexahydro-1H-4,12-methanobenzofuro[3,2-e]isoquinolin-7(7aH)-one, 1j) showed ∼75 times better TLR-4 antagonist activity than (+)-naltrexone, and the ratio of its cell viability IC50, a measure of its toxicity, to TLR-4 antagonist activity (140 μM/1.4 μM) was among the best of the new analogs. This compound (1j) was active in vivo; it significantly increased and prolonged morphine analgesia.
Collapse
Affiliation(s)
- Brandon R Selfridge
- †Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Xiaohui Wang
- ‡Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States.,§Department of Chemistry and Biochemistry the BioFrontiers Institute, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Yingning Zhang
- ‡Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Hang Yin
- §Department of Chemistry and Biochemistry the BioFrontiers Institute, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Peter M Grace
- ‡Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Linda R Watkins
- ‡Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Arthur E Jacobson
- †Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Kenner C Rice
- †Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| |
Collapse
|
124
|
Arout CA, Edens E, Petrakis IL, Sofuoglu M. Targeting Opioid-Induced Hyperalgesia in Clinical Treatment: Neurobiological Considerations. CNS Drugs 2015; 29:465-86. [PMID: 26142224 DOI: 10.1007/s40263-015-0255-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Opioid analgesics have become a cornerstone in the treatment of moderate to severe pain, resulting in a steady rise of opioid prescriptions. Subsequently, there has been a striking increase in the number of opioid-dependent individuals, opioid-related overdoses, and fatalities. Clinical use of opioids is further complicated by an increasingly deleterious profile of side effects beyond addiction, including tolerance and opioid-induced hyperalgesia (OIH), where OIH is defined as an increased sensitivity to already painful stimuli. This paradoxical state of increased nociception results from acute and long-term exposure to opioids, and appears to develop in a substantial subset of patients using opioids. Recently, there has been considerable interest in developing an efficacious treatment regimen for acute and chronic pain. However, there are currently no well-established treatments for OIH. Several substrates have emerged as potential modulators of OIH, including the N-methyl-D-aspartate and γ-aminobutyric acid receptors, and most notably, the innate neuroimmune system. This review summarizes the neurobiology of OIH in the context of clinical treatment; specifically, we review evidence for several pathways that show promise for the treatment of pain going forward, as prospective adjuvants to opioid analgesics. Overall, we suggest that this paradoxical state be considered an additional target of clinical treatment for chronic pain.
Collapse
Affiliation(s)
- Caroline A Arout
- Department of Psychiatry, Yale University School of Medicine, VA Connecticut Healthcare System, 950 Campbell Avenue, West Haven, CT, 06516, USA,
| | | | | | | |
Collapse
|
125
|
Discovery of a novel site of opioid action at the innate immune pattern-recognition receptor TLR4 and its role in addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 118:129-63. [PMID: 25175864 DOI: 10.1016/b978-0-12-801284-0.00006-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Opioids have historically, and continue to be, an integral component of pain management. However, despite pharmacokinetic and dynamic optimization over the past 100 years, opioids continue to produce many undesirable side effects such as tolerance, reward, and dependence. As such, opioids are liable for addiction. Traditionally, opioid addiction was viewed as a solely neuronal process, and while substantial headway has been made into understanding the molecular and cellular mechanisms mediating this process, research has however, been relatively ambivalent to how the rest of the central nervous system (CNS) responds to opioids. Evidence over the past 20 years has clearly demonstrated the importance of the immunocompetent cells of the CNS (glia) in many aspects of opioid pharmacology. Particular focus has been placed on microglia and astrocytes, who in response to opioids, become activated and release inflammatory mediators. Importantly, the mechanism underlying immune activation is beginning to be elucidated. Evidence suggests an innate immune pattern-recognition receptor (toll-like receptor 4) as an integral component underlying opioid-induced glial activation. The subsequent proinflammatory response may be viewed akin to neurotransmission creating a process termed central immune signaling. Translationally, we are beginning to appreciate the importance of central immune signaling as it contributes to many behavioral actions of addiction including reward, withdrawal, and craving. As such, the aim of this chapter is to review and integrate the neuronal and central immune signaling perspective of addiction.
Collapse
|
126
|
Stolyarova A, Thompson AB, Barrientos RM, Izquierdo A. Reductions in frontocortical cytokine levels are associated with long-lasting alterations in reward valuation after methamphetamine. Neuropsychopharmacology 2015; 40:1234-42. [PMID: 25409594 PMCID: PMC4367468 DOI: 10.1038/npp.2014.309] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/11/2014] [Accepted: 11/13/2014] [Indexed: 12/11/2022]
Abstract
Alterations in reward valuation are thought to have a central role at all stages of the addiction process. We previously reported work aversion in an effortful T-maze task following a binge exposure to methamphetamine, and no such changes in effort following escalating doses. Limitations of the T-maze task include its two available options, with an effort requirement, in the form of increasing barrier height, varying incrementally as a function of time, and reward magnitudes held constant. Reward preferences and choices, however, are likely affected by the number of options available and the manner in which alternatives are presented. In the present experiment, we investigated the long-lasting, off-drug effects of methamphetamine on reward choices in a novel effortful maze task with three possible courses of action, each associated with different effort requirements and reward magnitudes. Neuroinflammatory responses associated with drug exposure, proposed as one of the mechanisms contributing to suboptimal choices on effort-based tasks, were also examined. We investigated region-specific changes in pro- and anti-inflammatory markers in the mesocorticolimbic pathway after methamphetamine, and their relationship with animals' reward choices. We observed long-lasting, increased sensitivity to differences in reward magnitude in the methamphetamine group: animals were more likely to overcome greater effort costs to obtain larger rewards on our novel effortful maze task. These behavioral changes were strongly predicted by pronounced decreases in frontocortical cytokines, but not amygdalar or striatal markers. The present results provide the first evidence that neuroinflammatory processes are associated with alterations in reward valuation during protracted drug withdrawal.
Collapse
Affiliation(s)
- Alexandra Stolyarova
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Andrew B Thompson
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ruth M Barrientos
- Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Alicia Izquierdo
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA,Department of Psychology, University of California, Los Angeles, 1285 Franz Hall Box 951563, Los Angeles, CA 90095-1563, USA, Tel: +1 310 825 3459, Fax: +1 310 206 5895, E-mail:
| |
Collapse
|
127
|
Montesinos J, Pascual M, Pla A, Maldonado C, Rodríguez-Arias M, Miñarro J, Guerri C. TLR4 elimination prevents synaptic and myelin alterations and long-term cognitive dysfunctions in adolescent mice with intermittent ethanol treatment. Brain Behav Immun 2015; 45:233-44. [PMID: 25486089 DOI: 10.1016/j.bbi.2014.11.015] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/29/2014] [Accepted: 11/26/2014] [Indexed: 12/30/2022] Open
Abstract
The adolescent brain undergoes important dynamic and plastic cell changes, including overproduction of axons and synapses, followed by rapid pruning along with ongoing axon myelination. These developmental changes make the adolescent brain particularly vulnerable to neurotoxic and behavioral effects of alcohol. Although the mechanisms of these effects are largely unknown, we demonstrated that ethanol by activating innate immune receptors toll-like receptor 4 (TLR4), induces neuroinflammation and brain damage in adult mice. The present study aims to evaluate whether intermittent ethanol treatment in adolescence promotes TLR4-dependent pro-inflammatory processes, leading to myelin and synaptic dysfunctions, and long-term cognitive impairments. Using wild-type (WT) and TLR4-deficient (TLR4-KO) adolescent mice treated intermittently with ethanol (3.0g/kg) for 2weeks, we show that binge-like ethanol treatment activates TLR4 signaling pathways (MAPK, NFκB) leading to the up-regulation of cytokines and pro-inflammatory mediators (COX-2, iNOS, HMGB1), impairing synaptic and myelin protein levels and causing ultrastructural alterations. These changes were associated with long-lasting cognitive dysfunctions in young adult mice, as demonstrated with the object recognition, passive avoidance and olfactory behavior tests. Notably, elimination of TLR4 receptors prevented neuroinflammation along with synaptic and myelin derangements, as well as long-term cognitive alterations. These results support the role of the neuroimmune response and TLR4 signaling in the neurotoxic and behavioral effects of ethanol in adolescence.
Collapse
Affiliation(s)
- Jorge Montesinos
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - María Pascual
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - Antoni Pla
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - Concepción Maldonado
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Jose Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain.
| |
Collapse
|
128
|
Pedraz M, Araos P, García-Marchena N, Serrano A, Romero-Sanchiz P, Suárez J, Castilla-Ortega E, Mayoral-Cleries F, Ruiz JJ, Pastor A, Barrios V, Chowen JA, Argente J, Torrens M, de la Torre R, Rodríguez De Fonseca F, Pavón FJ. Sex differences in psychiatric comorbidity and plasma biomarkers for cocaine addiction in abstinent cocaine-addicted subjects in outpatient settings. Front Psychiatry 2015; 6:17. [PMID: 25762940 PMCID: PMC4329735 DOI: 10.3389/fpsyt.2015.00017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/29/2015] [Indexed: 12/15/2022] Open
Abstract
There are sex differences in the progression of drug addiction, relapse, and response to therapies. Because biological factors participate in these differences, they should be considered when using biomarkers for addiction. In the current study, we evaluated the sex differences in psychiatric comorbidity and the concentrations of plasma mediators that have been reported to be affected by cocaine. Fifty-five abstinent cocaine-addicted subjects diagnosed with lifetime cocaine use disorders (40 men and 15 women) and 73 healthy controls (48 men and 25 women) were clinically assessed with the diagnostic interview "Psychiatric Research Interview for Substance and Mental Disorders." Plasma concentrations of chemokines, cytokines, N-acyl-ethanolamines, and 2-acyl-glycerols were analyzed according to history of cocaine addiction and sex, controlling for covariates age and body mass index (BMI). Relationships between these concentrations and variables related to cocaine addiction were also analyzed in addicted subjects. The results showed that the concentrations of chemokine (C-C motif) ligand 2/monocyte chemotactic protein-1 (CCL2/MCP-1) and chemokine (C-X-C motif) ligand 12/stromal cell-derived factor-1 (CXCL12/SDF-1) were only affected by history of cocaine addiction. The plasma concentrations of interleukin 1-beta (IL-1β), IL-6, IL-10, and tumor necrosis factor-alpha (TNFα) were affected by history of cocaine addiction and sex. In fact, whereas cytokine concentrations were higher in control women relative to men, these concentrations were reduced in cocaine-addicted women without changes in addicted men. Regarding fatty acid derivatives, history of cocaine addiction had a main effect on the concentration of each acyl derivative, whereas N-acyl-ethanolamines were increased overall in the cocaine group, 2-acyl-glycerols were decreased. Interestingly, N-palmitoleoyl-ethanolamine (POEA) was only increased in cocaine-addicted women. The covariate BMI had a significant effect on POEA and N-arachidonoyl-ethanolamine concentrations. Regarding psychiatric comorbidity in the cocaine group, women had lower incidence rates of comorbid substance use disorders than did men. For example, alcohol use disorders were found in 80% of men and 40% of women. In contrast, the addicted women had increased prevalences of comorbid psychiatric disorders (i.e., mood, anxiety, and psychosis disorders). Additionally, cocaine-addicted subjects showed a relationship between the concentrations of N-stearoyl-ethanolamine and 2-linoleoyl-glycerol and diagnosis of psychiatric comorbidity. These results demonstrate the existence of a sex influence on plasma biomarkers for cocaine addiction and on the presence of comorbid psychopathologies for clinical purposes.
Collapse
Affiliation(s)
- María Pedraz
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Pedro Araos
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Nuria García-Marchena
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Antonia Serrano
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad 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, Universidad de Málaga, Málaga, Spain
| | - Juan Suárez
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad 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, Universidad de Málaga, Málaga, Spain
| | - Fermín Mayoral-Cleries
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Juan Jesús Ruiz
- Centro Provincial de Drogodependencia, Diputación de Málaga, Málaga, Spain
| | - Antoni Pastor
- Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Barcelona, Spain
- Facultat de Medicina, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Vicente Barrios
- Department of Pediatrics and Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Julie A. Chowen
- Department of Pediatrics and Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Pediatrics and Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Torrens
- Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Barcelona, Spain
- Facultat de Medicina, Universitat Autonoma de Barcelona, Barcelona, Spain
- Institut de Neuropsiquiatria i Addiccions (INAD) del Parc de Salut MAR, Barcelona, Spain
| | - Rafael de la Torre
- Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Facultat de Ciencies de la Salut i de la Vida, Universitat Pompeu Fabra (CEXS-UPF), Barcelona, Spain
| | - Fernando Rodríguez De Fonseca
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Javier Pavón
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| |
Collapse
|
129
|
Zwicker JD, Zhang Y, Ren J, Hutchinson MR, Rice KC, Watkins LR, Greer JJ, Funk GD. Glial TLR4 signaling does not contribute to opioid-induced depression of respiration. J Appl Physiol (1985) 2014; 117:857-68. [PMID: 25103966 DOI: 10.1152/japplphysiol.00534.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Opioids activate glia in the central nervous system in part by activating the toll-like receptor 4 (TLR4)/myeloid differentiation 2 (MD2) complex. TLR4/MD2-mediated activation of glia by opioids compromises their analgesic actions. Glial activation is also hypothesized as pivotal in opioid-mediated reward and tolerance and as a contributor to opioid-mediated respiratory depression. We tested the contribution of TLR4 to opioid-induced respiratory depression using rhythmically active medullary slices that contain the pre-Bötzinger Complex (preBötC, an important site of respiratory rhythm generation) and adult rats in vivo. Injection with DAMGO (μ-opioid receptor agonist; 50 μM) or bath application of DAMGO (500 nM) or fentanyl (1 μM) slowed frequency recorded from XII nerves to 40%, 40%, or 50% of control, respectively. This DAMGO-mediated frequency inhibition was unaffected by preapplication of lipopolysaccharides from Rhodobacter sphaeroides (a TLR4 antagonist, 2,000 ng/ml) or (+)naloxone (1-10 μM, a TLR4-antagonist). Bath application of (-)naloxone (500 nM; a TLR4 and μ-opioid antagonist), however, rapidly reversed the opioid-mediated frequency decrease. We also compared the opioid-induced respiratory depression in slices in vitro in the absence and presence of bath-applied minocycline (an inhibitor of microglial activation) and in slices prepared from mice injected (ip) 18 h earlier with minocycline or saline. Minocycline had no effect on respiratory depression in vitro. Finally, the respiratory depression evoked in anesthetized rats by tail vein infusion of fentanyl was unaffected by subsequent injection of (+)naloxone, but completely reversed by (-)naloxone. These data indicate that neither activation of microglia in preBötC nor TLR4/MD2-activation contribute to opioid-induced respiratory depression.
Collapse
Affiliation(s)
- Jennifer D Zwicker
- Department of Physiology, Neuroscience and Mental Health Institute, Women and Children's Health Research Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Yong Zhang
- Department of Physiology, Neuroscience and Mental Health Institute, Women and Children's Health Research Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jun Ren
- Department of Physiology, Neuroscience and Mental Health Institute, Women and Children's Health Research Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mark R Hutchinson
- Discipline of Physiology, School of Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Kenner C Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Rockville, Maryland; and
| | - Linda R Watkins
- Department of Psychology and The Center for Neuroscience, University of Colorado at Boulder, Boulder, Colorado
| | - John J Greer
- Department of Physiology, Neuroscience and Mental Health Institute, Women and Children's Health Research Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Gregory D Funk
- Department of Physiology, Neuroscience and Mental Health Institute, Women and Children's Health Research Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada;
| |
Collapse
|
130
|
Reece AS, Hulse GK. Impact of lifetime opioid exposure on arterial stiffness and vascular age: cross-sectional and longitudinal studies in men and women. BMJ Open 2014; 4:e004521. [PMID: 24889849 PMCID: PMC4054659 DOI: 10.1136/bmjopen-2013-004521] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE To characterise and compare the potentiation of arterial stiffness and vascular ageing by opioids in men and women. DESIGN Cross-sectional and longitudinal studies of 576 clinical controls and 687 opioid-dependent patients (ODP) on 710 and 1305 occasions, respectively, over a total of 2382 days (6.52 years), 2006-2011. Methodology Radial pulse wave analysis with Atcor SphygmoCor system (Sydney). SETTING Primary care. CONTROLS General practice patients with non-cardiovascular disorders, and university student controls. ODP: Patients undergoing clinical management of their opioid dependence. CONTROLS had lower chronological ages (CAs) than ODP (30.0±0.5 vs 34.5±0.3, mean±SEM, p<0.0001). 69.6% and 67.7% participants were men, and 16% and 92.3% were smokers (p<0.0001) for controls and ODP, respectively. 86.3%, 10.3% and 3.4% of ODP were treated with buprenorphine (6.98±0.21 mg), methadone (63.04±4.01 mg) or implant naltrexone, respectively. Body mass index (BMI) was depressed in ODP. INTERVENTIONS Nil. PRIMARY OUTCOME MEASURES Vascular Reference Age (RA) and the ratio of vascular age to chronological age (RA/CA). SECONDARY OUTCOME MEASURES Arterial stiffness including Augmentation Index. RESULTS After BMI adjustment, RA in ODP was higher as a function of CA and of time (both p<0.05). Modelled mean RA in control and ODP was 35.6 and 36.3 years (+1.97%) in men, and 34.5 and 39.2 years (+13.43%) in women, respectively. Changes in RA and major arterial stiffness indices were worse in women both as a factor (p = 0.0036) and in interaction with CA (p = 0.0040). Quadratic, cubic and quartic functions of opioid exposure duration outperformed linear models with RA/CA over CA and over time. The opioid dose-response relationship persisted longitudinally after multiple adjustments from p=0.0013 in men and p=0.0073 in women. CONCLUSIONS Data show that lifetime opioid exposure, an interactive cardiovascular risk factor, particularly in women, is related to linear, quadratic, cubic and quartic functions of treatment duration and is consistent with other literature of accelerated ageing in patients with OD.
Collapse
Affiliation(s)
- Albert Stuart Reece
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Gary Kenneth Hulse
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
131
|
Isaza C, Henao J, Velez J, Rodríguez MA, Sierra J, Beltrán L, Sepúlveda A. [Evaluation of the Methadone Maintenance Program of the Risaralda Mental Hospital]. ACTA ACUST UNITED AC 2014; 43:96-105. [PMID: 26574964 DOI: 10.1016/j.rcp.2014.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 02/01/2014] [Accepted: 02/04/2014] [Indexed: 11/17/2022]
Abstract
BACKGROUND Psychosocial care and methadone maintenance treatment (MMT) are the preferred strategies for the management of heroin addicts, but the results are still unsatisfactory, justifying the search and intervention of the factors influencing the response to treatment. METHODOLOGY In order to determine the contribution of demographic, clinical and genetic variables on serum concentrations and response to methadone, we investigated patients on MMT, who were receiving methadone in supervised and unchanged doses at least during the previous two weeks. The age, gender, body mass index (BMI), duration of heroin abuse, addiction to other drugs, criminal background, current daily methadone doses, time spent in the TMM, comorbidity and concomitant medication were recorded. Blood samples were taken for the determination of serum levels of racemic methadone and its R and S-enantiomers, and for typing of candidate alleles of POR, CYP2B6, ABCB1, GRIN1, OPRM1, SLC6A3, DßH and ARRB2 genes, all associated with the metabolism, tissue distribution and mechanism of action of methadone. Methadone quantification was by HPLC-DAD, and the detection of genetic markers by Real Time PCR and VNTR methods. RESULTS A total of 80 subject volunteers were enrolled, with a mean age of 23.5 (5) years (86% male), all of them were addicts of multiple drugs, 60% with a criminal background, 5.1 (2.9) years taking heroin, and 5.3 (4) months on MMT, and taking a supervised dose of 41 (12) mg/day methadone. The (R), (S) and (R, S) methadone enantiomer trough plasma levels were, 84 (40), 84 (42), and 168 (77) ng/mL, respectively. All genotypes were in Hardy-Weinberg equilibrium. The two urine tests were negative for heroin in 61.3% (49/80) of the volunteers, the decline in cocaine/crack use was 83%, 30% of marijuana, and other psychoactives (inhalants, benzodiazepines, amphetamines) decreased to zero, while the consumption of snuff remained at 93.5% (75/80). Blood concentrations of racemic methadone and its enantiomers were significantly associated with the dose/day of the medication, but none of the other demographic, clinical or genetic variables impacted on serum levels of methadone. As for the results of the MMT, non-users and occasional users of heroin, as well as those who stopped taking other psychoactive drugs, and the ones who did not, were similar as regards the demographic, genetic and clinical variables. This included the blood metahdone concentrations, except for individuals who did not reduce their consumption of other psychoactives other than heroin, who had significantly (P=.03) higher blood levels of S-methadone, compared with those who did stop taking them. CONCLUSIONS There was a significant reduction in the consumption of heroin and other psychoactives, and social rehabilitation of patients. However, the extensive overlap between effective and ineffective doses of methadone suggests the presence of personal and social variables that transcend the simple pharmacological management. These probably need to be addressed more successfully from the psychosocial features, particularly as regards to identifying and overcoming relapse-trigger experiences, as well as certain features of the patient, such as their psychological distress level or their psychiatric disorders.
Collapse
Affiliation(s)
- Carlos Isaza
- Farmacología, Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira, Colombia.
| | - Julieta Henao
- Genética, Facultad de Ciencias de la Salud, Laboratorio de Genética Médica, Universidad Tecnológica de Pereira, Pereira, Colombia
| | - Juan Velez
- Epidemiología, Dirección Científica, ESE Hospital Mental de Risaralda, Pereira, Colombia
| | | | - Juan Sierra
- Farmacodependencia, ESE Hospital Mental de Risaralda, Pereira, Colombia
| | - Leonardo Beltrán
- Laboratorio de Genética Médica, Universidad Tecnológica de Pereira, Pereira, Colombia
| | - Ana Sepúlveda
- Laboratorio de Genética Médica, Universidad Tecnológica de Pereira, Pereira, Colombia
| |
Collapse
|
132
|
Hauser KF, Knapp PE. Interactions of HIV and drugs of abuse: the importance of glia, neural progenitors, and host genetic factors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 118:231-313. [PMID: 25175867 PMCID: PMC4304845 DOI: 10.1016/b978-0-12-801284-0.00009-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Considerable insight has been gained into the comorbid, interactive effects of HIV and drug abuse in the brain using experimental models. This review, which considers opiates, methamphetamine, and cocaine, emphasizes the importance of host genetics and glial plasticity in driving the pathogenic neuron remodeling underlying neuro-acquired immunodeficiency syndrome and drug abuse comorbidity. Clinical findings are less concordant than experimental work, and the response of individuals to HIV and to drug abuse can vary tremendously. Host-genetic variability is important in determining viral tropism, neuropathogenesis, drug responses, and addictive behavior. However, genetic differences alone cannot account for individual variability in the brain "connectome." Environment and experience are critical determinants in the evolution of synaptic circuitry throughout life. Neurons and glia both exercise control over determinants of synaptic plasticity that are disrupted by HIV and drug abuse. Perivascular macrophages, microglia, and to a lesser extent astroglia can harbor the infection. Uninfected bystanders, especially astroglia, propagate and amplify inflammatory signals. Drug abuse by itself derails neuronal and glial function, and the outcome of chronic exposure is maladaptive plasticity. The negative consequences of coexposure to HIV and drug abuse are determined by numerous factors including genetics, sex, age, and multidrug exposure. Glia and some neurons are generated throughout life, and their progenitors appear to be targets of HIV and opiates/psychostimulants. The chronic nature of HIV and drug abuse appears to result in sustained alterations in the maturation and fate of neural progenitors, which may affect the balance of glial populations within multiple brain regions.
Collapse
Affiliation(s)
- Kurt F Hauser
- Department of Pharmacology & Toxicology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA.
| | - Pamela E Knapp
- Department of Pharmacology & Toxicology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA; Department of Anatomy & Neurobiology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
133
|
Cui C, Shurtleff D, Harris RA. Neuroimmune mechanisms of alcohol and drug addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 118:1-12. [PMID: 25175859 DOI: 10.1016/b978-0-12-801284-0.00001-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alcohol and other drugs of abuse have significant impacts on the neuroimmune system. Studies have demonstrated that drugs of abuse interact with the neuroimmune system and alter neuroimmune gene expression and signaling, which in turn contribute to various aspects of addiction. As the key component of the CNS immune system, neuroimmune factors mediate neuroinflammation and modulate a wide range of brain function including neuronal activity, endocrine function, and CNS development. These neuromodulatory properties of immune factors, together with their essential role in neuroinflammation, provide a new framework to understand neuroimmune mechanisms mediating brain functional and behavioral changes contributing to addiction. This chapter highlights recent advances in understanding neuroimmune changes associated with exposure to alcohol and other drugs of abuse, including opiates, marijuana, methamphetamine, and cocaine. It provides a brief overview on what we know about neuroimmune signaling and its role in drug action and addiction.
Collapse
Affiliation(s)
- Changhai Cui
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA.
| | - David Shurtleff
- National Center for Complementary & Alternative Medicine, Bethesda, Maryland, USA
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
134
|
Beardsley PM, Hauser KF. Glial modulators as potential treatments of psychostimulant abuse. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:1-69. [PMID: 24484974 DOI: 10.1016/b978-0-12-420118-7.00001-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Glia (including astrocytes, microglia, and oligodendrocytes), which constitute the majority of cells in the brain, have many of the same receptors as neurons, secrete neurotransmitters and neurotrophic and neuroinflammatory factors, control clearance of neurotransmitters from synaptic clefts, and are intimately involved in synaptic plasticity. Despite their prevalence and spectrum of functions, appreciation of their potential general importance has been elusive since their identification in the mid-1800s, and only relatively recently have they been gaining their due respect. This development of appreciation has been nurtured by the growing awareness that drugs of abuse, including the psychostimulants, affect glial activity, and glial activity, in turn, has been found to modulate the effects of the psychostimulants. This developing awareness has begun to illuminate novel pharmacotherapeutic targets for treating psychostimulant abuse, for which targeting more conventional neuronal targets has not yet resulted in a single, approved medication. In this chapter, we discuss the molecular pharmacology, physiology, and functional relationships that the glia have especially in the light in which they present themselves as targets for pharmacotherapeutics intended to treat psychostimulant abuse disorders. We then review a cross section of preclinical studies that have manipulated glial processes whose behavioral effects have been supportive of considering the glia as drug targets for psychostimulant-abuse medications. We then close with comments regarding the current clinical evaluation of relevant compounds for treating psychostimulant abuse, as well as the likelihood of future prospects.
Collapse
Affiliation(s)
| | - Kurt F Hauser
- Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
135
|
Hagen EH, Roulette CJ, Sullivan RJ. Explaining human recreational use of 'pesticides': The neurotoxin regulation model of substance use vs. the hijack model and implications for age and sex differences in drug consumption. Front Psychiatry 2013; 4:142. [PMID: 24204348 PMCID: PMC3817850 DOI: 10.3389/fpsyt.2013.00142] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 10/12/2013] [Indexed: 12/21/2022] Open
Abstract
Most globally popular drugs are plant neurotoxins or their close chemical analogs. These compounds evolved to deter, not reward or reinforce, consumption. Moreover, they reliably activate virtually all toxin defense mechanisms, and are thus correctly identified by human neurophysiology as toxins. Acute drug toxicity must therefore play a more central role in drug use theory. We accordingly challenge the popular idea that the rewarding and reinforcing properties of drugs "hijack" the brain, and propose instead that the brain evolved to carefully regulate neurotoxin consumption to minimize fitness costs and maximize fitness benefits. This perspective provides a compelling explanation for the dramatic changes in substance use that occur during the transition from childhood to adulthood, and for pervasive sex differences in substance use: because nicotine and many other plant neurotoxins are teratogenic, children, and to a lesser extent women of childbearing age, evolved to avoid ingesting them. However, during the course of human evolution many adolescents and adults reaped net benefits from regulated intake of plant neurotoxins.
Collapse
Affiliation(s)
- Edward H. Hagen
- Department of Anthropology, Washington State University, Vancouver, WA, USA
| | - Casey J. Roulette
- Department of Anthropology, Washington State University, Vancouver, WA, USA
| | - Roger J. Sullivan
- Department of Anthropology, California State University, Sacramento, CA, USA
| |
Collapse
|
136
|
Campbell LA, Avdoshina V, Rozzi S, Mocchetti I. CCL5 and cytokine expression in the rat brain: differential modulation by chronic morphine and morphine withdrawal. Brain Behav Immun 2013; 34:130-40. [PMID: 23968971 PMCID: PMC3795805 DOI: 10.1016/j.bbi.2013.08.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/12/2013] [Accepted: 08/13/2013] [Indexed: 01/23/2023] Open
Abstract
Opioids have been shown to influence the immune system and to promote the expression of pro-inflammatory cytokines in the central nervous system. However, recent data have shown that activation of opioid receptors increases the expression and release of the neuroprotective chemokine CCL5 from astrocytes in vitro. To further define the interaction between CCL5 and inflammation in response to opioids, we have examined the effect of chronic morphine and morphine withdrawal on the in vivo expression of CCL5 as well as of pro-inflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). Rats undergoing a chronic morphine paradigm (10 mg/kg increasing to 30 mg/kg, twice a day for 5 days) showed a twofold increase of CCL5 protein and mRNA within the cortex and striatum. No changes were observed in the levels of IL-1β and TNF-α. Naltrexone blocked the effect of morphine. A chronic morphine paradigm with no escalating doses (10 mg/kg, twice a day) did not alter CCL5 levels compared to saline-treated animals. On the contrary, rats undergoing spontaneous morphine withdrawal exhibited lower levels of CCL5 within the cortex as well as increased levels of pro-inflammatory cytokines and Iba-1 positive cells than saline-treated rats. Overall, these data suggest that morphine withdrawal may promote cytokines and other inflammatory responses that have the potential of exacerbating neuronal damage.
Collapse
Affiliation(s)
- Lee A. Campbell
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC 20057
| | - Valeriya Avdoshina
- Department of Neuroscience, Georgetown University Medical Center, Washington DC 20057
| | - Summer Rozzi
- Interdisciplinary Program of Neuroscience, Laboratory of Preclinical Neurobiology, Georgetown University Medical Center, Washington DC 20057
| | - Italo Mocchetti
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC 20057,Department of Neuroscience, Georgetown University Medical Center, Washington DC 20057
| |
Collapse
|
137
|
Doyon WM, Thomas AM, Ostroumov A, Dong Y, Dani JA. Potential substrates for nicotine and alcohol interactions: a focus on the mesocorticolimbic dopamine system. Biochem Pharmacol 2013; 86:1181-93. [PMID: 23876345 DOI: 10.1016/j.bcp.2013.07.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 07/11/2013] [Accepted: 07/12/2013] [Indexed: 01/13/2023]
Abstract
Epidemiological studies consistently find correlations between nicotine and alcohol use, yet the neural mechanisms underlying their interaction remain largely unknown. Nicotine and alcohol (i.e., ethanol) share many common molecular and cellular targets that provide potential substrates for nicotine-alcohol interactions. These targets for interaction often converge upon the mesocorticolimbic dopamine system, where the link to drug self-administration and reinforcement is well documented. Both nicotine and alcohol activate the mesocorticolimbic dopamine system, producing downstream dopamine signals that promote the drug reinforcement process. While nicotine primarily acts via nicotinic acetylcholine receptors, alcohol acts upon a wider range of receptors and molecular substrates. The complex pharmacological profile of these two drugs generates overlapping responses that ultimately intersect within the mesocorticolimbic dopamine system to promote drug use. Here we will examine overlapping targets between nicotine and alcohol and provide evidence for their interaction. Based on the existing literature, we will also propose some potential targets that have yet to be directly tested. Mechanistic studies that examine nicotine-alcohol interactions would ultimately improve our understanding of the factors that contribute to the associations between nicotine and alcohol use.
Collapse
Affiliation(s)
- William M Doyon
- Center on Addiction, Learning, Memory, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
138
|
Hutchinson MR, Watkins LR. Why is neuroimmunopharmacology crucial for the future of addiction research? Neuropharmacology 2013; 76 Pt B:218-27. [PMID: 23764149 DOI: 10.1016/j.neuropharm.2013.05.039] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 05/13/2013] [Accepted: 05/23/2013] [Indexed: 12/13/2022]
Abstract
A major development in drug addiction research in recent years has been the discovery that immune signaling within the central nervous system contributes significantly to mesolimbic dopamine reward signaling induced by drugs of abuse, and hence is involved in the presentation of reward behaviors. Additionally, in the case of opioids, these hypotheses have advanced through to the discovery of the novel site of opioid action at the innate immune pattern recognition receptor Toll-like receptor 4 as the necessary triggering event that engages this reward facilitating central immune signaling. Thus, the hypothesis of major proinflammatory contributions to drug abuse was born. This review will examine these key discoveries, but also address several key lingering questions of how central immune signaling is able to contribute in this fashion to the pharmacodynamics of drugs of abuse. It is hoped that by combining the collective wisdom of neuroscience, immunology and pharmacology, into Neuroimmunopharmacology, we may more fully understanding the neuronal and immune complexities of how drugs of abuse, such as opioids, create their rewarding and addiction states. Such discoveries will point us in the direction such that one day soon we might successfully intervene to successfully treat drug addiction. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Mark R Hutchinson
- Discipline of Physiology, School of Medical Sciences, University of Adelaide, Level 5, Medical School South, Frome Rd, Adelaide, South Australia 5005, Australia.
| | | |
Collapse
|
139
|
Loftis JM, Huckans M. Substance use disorders: psychoneuroimmunological mechanisms and new targets for therapy. Pharmacol Ther 2013; 139:289-300. [PMID: 23631821 DOI: 10.1016/j.pharmthera.2013.04.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/15/2013] [Indexed: 12/28/2022]
Abstract
An estimated 76.4 million people worldwide meet criteria for alcohol use disorders, and 15.3 million meet criteria for drug use disorders. Given the high rates of addiction and the associated health, economic, and social costs, it is essential to develop a thorough understanding of the impact of substance abuse on mental and physical health outcomes and to identify new treatment approaches for substance use disorders (SUDs). Psychoneuroimmunology is a rapidly expanding, multidisciplinary area of research that may be of particular importance to addiction medicine, as its focus is on the dynamic and complex interactions among behavioral factors, the central nervous system, and the endocrine and immune systems (Ader, 2001). This review, therefore, focuses on: 1) the psychoneuroimmunologic effects of SUDs by substance type and use pattern, and 2) the current and future treatment strategies, including barriers that can impede successful recovery outcomes. Evidence-based psychosocial and pharmacotherapeutic treatments are reviewed. Psychological factors and central nervous system correlates that impact treatment adherence and response are discussed. Several novel therapeutic approaches that are currently under investigation are introduced; translational data from animal and human studies is presented, highlighting immunotherapy as a promising new direction for addiction medicine.
Collapse
Affiliation(s)
- Jennifer M Loftis
- Research and Development Service, Portland VA Medical Center, Portland, OR, USA.
| | | |
Collapse
|
140
|
Theberge FR, Li X, Kambhampati S, Pickens CL, St Laurent R, Bossert JM, Baumann MH, Hutchinson MR, Rice KC, Watkins LR, Shaham Y. Effect of chronic delivery of the Toll-like receptor 4 antagonist (+)-naltrexone on incubation of heroin craving. Biol Psychiatry 2013; 73:729-37. [PMID: 23384483 PMCID: PMC3615146 DOI: 10.1016/j.biopsych.2012.12.019] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 12/10/2012] [Accepted: 12/27/2012] [Indexed: 11/18/2022]
Abstract
BACKGROUND Recent evidence implicates toll-like receptor 4 (TLR4) in opioid analgesia, tolerance, conditioned place preference, and self-administration. Here, we determined the effect of the TLR4 antagonist (+)-naltrexone (a μ-opioid receptor inactive isomer) on the time-dependent increases in cue-induced heroin seeking after withdrawal (incubation of heroin craving). METHODS In an initial experiment, we trained rats for 9 hours per day to self-administer heroin (.1 mg/kg/infusion) for 9 days; lever presses were paired with a 5-second tone-light cue. We then assessed cue-induced heroin seeking in 30-minute extinction sessions on withdrawal day 1; immediately after testing, we surgically implanted rats with Alzet minipumps delivering (+)-naltrexone (0, 7.5, 15, 30 mg/kg/day, subcutaneous) for 14 days. We then tested the rats for incubated cue-induced heroin seeking in 3-hour extinction tests on withdrawal day 13. RESULTS We found that chronic delivery of (+)-naltrexone via minipumps during the withdrawal phase decreased incubated cue-induced heroin seeking. In follow-up experiments, we found that acute injections of (+)-naltrexone immediately before withdrawal day 13 extinction tests had no effect on incubated cue-induced heroin seeking. Furthermore, chronic delivery of (+)-naltrexone (15 or 30 mg/kg/day) or acute systemic injections (15 or 30 mg/kg) had no effect on ongoing extended access heroin self-administration. Finally, in rats trained to self-administer methamphetamine (.1 mg/kg/infusion, 9 hours/day, 9 days), chronic delivery of (+)-naltrexone (30 mg/kg/day) during the withdrawal phase had no effect on incubated cue-induced methamphetamine seeking. CONCLUSIONS The present results suggest a critical role of TLR4 in the development of incubation of heroin, but not methamphetamine, craving.
Collapse
Affiliation(s)
- Florence R Theberge
- Intramural Research Program of the National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Crews FT, Qin L, Sheedy D, Vetreno RP, Zou J. High mobility group box 1/Toll-like receptor danger signaling increases brain neuroimmune activation in alcohol dependence. Biol Psychiatry 2013; 73:602-12. [PMID: 23206318 PMCID: PMC3602398 DOI: 10.1016/j.biopsych.2012.09.030] [Citation(s) in RCA: 229] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 08/30/2012] [Accepted: 09/19/2012] [Indexed: 12/30/2022]
Abstract
BACKGROUND Innate immune gene expression is regulated in part through high mobility group box 1 (HMGB1), an endogenous proinflammatory cytokine, that activates multiple members of the interleukin-1/Toll-like receptor (TLR) family associated with danger signaling. We investigated expression of HMGB1, TLR2, TLR3, and TLR4 in chronic ethanol-treated mouse brain, postmortem human alcoholic brain, and rat brain slice culture to test the hypothesis that neuroimmune activation in alcoholic brain involves ethanol activation of HMGB1/TLR danger signaling. METHODS Protein levels were assessed using Western blot, enzyme-linked immunosorbent assay, and immunohistochemical immunoreactivity (+IR), and messenger RNA (mRNA) levels were measured by real time polymerase chain reaction in ethanol-treated mice (5 g/kg/day, intragastric, 10 days + 24 hours), rat brain slice culture, and postmortem human alcoholic brain. RESULTS Ethanol treatment of mice increased brain mRNA and +IR protein expression of HMGB1, TLR2, TLR3, and TLR4. Postmortem human alcoholic brain also showed increased HMGB1, TLR2, TLR3, and TLR4 +IR cells that correlated with lifetime alcohol consumption, as well as each other. Ethanol treatment of brain slice culture released HMGB1 into the media and induced the proinflammatory cytokine, interleukin-1 beta (IL-1β). Neutralizing antibodies to HMGB1 and small inhibitory mRNA to HMGB1 or TLR4 blunted ethanol induction of IL-1β. CONCLUSIONS Ethanol-induced HMGB1/TLR signaling contributes to induction of the proinflammatory cytokine, IL-1β. Increased expression of HMGB1, TLR2, TLR3, and TLR4 in alcoholic brain and in mice treated with ethanol suggests that chronic alcohol-induced brain neuroimmune activation occurs through HMGB1/TLR signaling.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies, Department of Pharmacology and Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7178, USA.
| | | | | | | | | |
Collapse
|
142
|
Neuroimmune signaling: a key component of alcohol abuse. Curr Opin Neurobiol 2013; 23:513-20. [PMID: 23434064 DOI: 10.1016/j.conb.2013.01.024] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/16/2013] [Accepted: 01/24/2013] [Indexed: 12/13/2022]
Abstract
Molecular and behavioral studies corroborate a pivotal role for the innate immune system in mediating the acute and chronic effects of alcohol and support a neuroimmune hypothesis of alcohol addiction. Changes in expression of neuroimmune genes and microglial transcripts occur in postmortem brain from alcoholics and animals exposed to alcohol, and null mutant animals lacking certain innate immune genes show decreased alcohol-mediated responses. Many of the differentially expressed genes are part of the toll like receptor (TLR) signaling pathway and culminate in an increased expression of pro-inflammatory immune genes. Compounds known to inhibit inflammation, microglial activation, and neuroimmune gene expression have shown promising results in reducing alcohol-mediated behaviors in animal models, indicating that neuroimmune signaling pathways offer unexplored targets in the treatment of alcohol abuse.
Collapse
|
143
|
Hutchinson MR, Northcutt AL, Hiranita T, Wang X, Lewis SS, Thomas J, van Steeg K, Kopajtic TA, Loram LC, Sfregola C, Galer E, Miles NE, Bland ST, Amat J, Rozeske RR, Maslanik T, Chapman TR, Strand KA, Fleshner M, Bachtell RK, Somogyi AA, Yin H, Katz JL, Rice KC, Maier SF, Watkins LR. Opioid activation of toll-like receptor 4 contributes to drug reinforcement. J Neurosci 2012; 32:11187-200. [PMID: 22895704 PMCID: PMC3454463 DOI: 10.1523/jneurosci.0684-12.2012] [Citation(s) in RCA: 231] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Revised: 06/14/2012] [Accepted: 06/25/2012] [Indexed: 01/08/2023] Open
Abstract
Opioid action was thought to exert reinforcing effects solely via the initial agonism of opioid receptors. Here, we present evidence for an additional novel contributor to opioid reward: the innate immune pattern-recognition receptor, toll-like receptor 4 (TLR4), and its MyD88-dependent signaling. Blockade of TLR4/MD2 by administration of the nonopioid, unnatural isomer of naloxone, (+)-naloxone (rats), or two independent genetic knock-outs of MyD88-TLR4-dependent signaling (mice), suppressed opioid-induced conditioned place preference. (+)-Naloxone also reduced opioid (remifentanil) self-administration (rats), another commonly used behavioral measure of drug reward. Moreover, pharmacological blockade of morphine-TLR4/MD2 activity potently reduced morphine-induced elevations of extracellular dopamine in rat nucleus accumbens, a region critical for opioid reinforcement. Importantly, opioid-TLR4 actions are not a unidirectional influence on opioid pharmacodynamics, since TLR4(-/-) mice had reduced oxycodone-induced p38 and JNK phosphorylation, while displaying potentiated analgesia. Similar to our recent reports of morphine-TLR4/MD2 binding, here we provide a combination of in silico and biophysical data to support (+)-naloxone and remifentanil binding to TLR4/MD2. Collectively, these data indicate that the actions of opioids at classical opioid receptors, together with their newly identified TLR4/MD2 actions, affect the mesolimbic dopamine system that amplifies opioid-induced elevations in extracellular dopamine levels, therefore possibly explaining altered opioid reward behaviors. Thus, the discovery of TLR4/MD2 recognition of opioids as foreign xenobiotic substances adds to the existing hypothesized neuronal reinforcement mechanisms, identifies a new drug target in TLR4/MD2 for the treatment of addictions, and provides further evidence supporting a role for central proinflammatory immune signaling in drug reward.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/blood
- Analysis of Variance
- Animals
- Conditioning, Operant/drug effects
- Conditioning, Operant/physiology
- Dopamine/metabolism
- Dose-Response Relationship, Drug
- Drug Administration Routes
- Hyperalgesia/drug therapy
- Hyperalgesia/physiopathology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Microdialysis
- Mitogen-Activated Protein Kinase 1/metabolism
- Models, Molecular
- Myeloid Differentiation Factor 88/deficiency
- Naloxone/pharmacology
- Narcotic Antagonists/pharmacology
- Nucleus Accumbens/drug effects
- Nucleus Accumbens/metabolism
- Pain Threshold/drug effects
- Pain Threshold/physiology
- Phosphorylation/drug effects
- Protein Binding/drug effects
- Protein Binding/genetics
- Rats
- Rats, Sprague-Dawley
- Reaction Time/drug effects
- Reinforcement, Psychology
- Self Administration
- Signal Transduction/drug effects
- Time Factors
- Toll-Like Receptor 4/agonists
- Toll-Like Receptor 4/deficiency
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
- M R Hutchinson
- Department of Psychology and Neuroscience, University of Colorado-Boulder, Boulder, Colorado 80309, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Kovács KJ. Microglia and drug-induced plasticity in reward-related neuronal circuits. Front Mol Neurosci 2012; 5:74. [PMID: 22707932 PMCID: PMC3374147 DOI: 10.3389/fnmol.2012.00074] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 05/25/2012] [Indexed: 12/16/2022] Open
Affiliation(s)
- Krisztina J Kovács
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine Budapest, Hungary
| |
Collapse
|