1
|
Luján MÁ, Kim Y, Zhang LY, Cheer JF. Cannabinoid-based Pharmacology for the Management of Substance Use Disorders. Curr Top Behav Neurosci 2025. [PMID: 39813001 DOI: 10.1007/7854_2024_551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
In the last two decades, the endocannabinoid system has emerged as a crucial modulator of motivation and emotional processing. Due to its widespread neuroanatomical distribution and characteristic retrograde signaling nature, cannabinoid type I receptors and their endogenous ligands finely orchestrate somatic and axon terminal activity of dopamine neurons. Owing to these unique features, this signaling system is a promising pharmacological target to ameliorate dopamine-mediated drug-seeking behaviors while circumventing the adverse side effects of, for instance, dopaminergic antagonists. Despite considerable preclinical efforts, an agreement on the efficacy of endocannabinoid-targeting compounds for treating drug substance use disorders in humans has not been reached. In the following chapter, we will summarize preclinical and clinical evidence addressing the therapeutic potential of cannabinoids and endocannabinoid-targeting compounds in substance use disorders. To bridge the gap between animal and clinical research, we capitalize on studies evaluating the impact of endocannabinoid-targeting compounds in relevant settings, such as the management of drug relapse. Finally, we discuss the therapeutic potential of novel cannabinoid compounds that hold promise for treating substance use disorders.
Collapse
Affiliation(s)
- M Á Luján
- Department of Neurobiology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Y Kim
- Department of Neurobiology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - L Y Zhang
- Department of Neurobiology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - J F Cheer
- Department of Neurobiology, University of Maryland, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Castro EM, Lotfipour S, Leslie FM. Neuroglia in substance use disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:347-369. [PMID: 40148055 DOI: 10.1016/b978-0-443-19102-2.00014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Substance use disorders (SUD) remain a major public health concern in which individuals are unable to control their use of substances despite significant harm and negative consequences. Drugs of abuse dysregulate major brain and behavioral functions. Glial cells, primarily microglia and astrocytes, play a crucial role in these drug-induced molecular and behavioral changes. This review explores preclinical and clinical studies of how neuroglia and their associated neuroinflammatory responses contribute to SUD and reward-related properties. We evaluate preclinical and clinical evidence for targeting neuroglia as therapeutic interventions. In addition, we evaluate the literature on the gut microbiome and its role in SUD. Clinical treatments are most effective for reducing drug cravings, and some have yielded promising results in other measures of drug use. N-Acetylcysteine, through modulation of cysteine-glutamate antiporter of glial cells, shows encouraging results across a variety of drug classes. Neuroglia and gut microbiome interactions are important factors to consider with regard to SUD and could lead to novel therapeutic avenues. Age- and sex-dependent properties of neuroglia, gut microbiome, and drug use behaviors are important areas in need of further investigation.
Collapse
Affiliation(s)
- Emily M Castro
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
| | - Shahrdad Lotfipour
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States; Department of Emergency Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States; Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Frances M Leslie
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States.
| |
Collapse
|
3
|
Cannella N, Lunerti V, Shen Q, Li H, Benvenuti F, Soverchia L, Narendran R, Weiss F, Ciccocioppo R. Cebranopadol, a novel long-acting opioid agonist with low abuse liability, to treat opioid use disorder: Preclinical evidence of efficacy. Neuropharmacology 2024; 257:110048. [PMID: 38901642 DOI: 10.1016/j.neuropharm.2024.110048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Maintenance therapy with buprenorphine and methadone is the gold standard pharmacological treatment for opioid use disorder (OUD). Despite these compounds demonstrating substantial efficacy, a significant number of patients do not show optimal therapeutic responses. The abuse liability of these medications is also a concern. Here we used rats to explore the therapeutic potential of the new long-acting pan-opioid agonist Cebranopadol in OUD. We tested the effect of cebranopadol on heroin self-administration and yohimbine-induced reinstatement of heroin seeking. In addition, we evaluated the abuse liability potential of cebranopadol in comparison to that of heroin under fixed ratio 1 (FR1) and progressive ratio (PR) operant self-administration contingencies. Oral administration of cebranopadol (0, 25, 50 μg/kg) significantly attenuated drug self-administration independent of heroin dose (1, 7, 20, 60μg/inf). Cebranopadol also reduced the break point for heroin (20 μg/inf). Finally, pretreatment with cebranopadol significantly attenuated yohimbine-induced reinstatement of drug seeking. In abuse liability experiments under FR1 contingency, rats maintained responding for heroin (1, 7, 20, 60μg/inf) to a larger extent than cebranopadol (0.03, 0.1, 0.3, 1.0, 6.0μg/inf). Under PR contingency, heroin maintained responding at high levels at all except the lowest dose, while the break point (BP) for cebranopadol did not differ from that of saline. Together, these data indicate that cebranopadol is highly efficacious in attenuating opioid self-administration and stress-induced reinstatement, while having limited abuse liability properties. Overall, the data suggest clinical potential of this compound for OUD treatment.
Collapse
Affiliation(s)
- Nazzareno Cannella
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy
| | - Veronica Lunerti
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy
| | - Qianwei Shen
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy
| | - Hongwu Li
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy; School of Chemical Engineering, Changchun University of Changchung, 130012, China
| | - Federica Benvenuti
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy
| | - Laura Soverchia
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Friedbert Weiss
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy.
| |
Collapse
|
4
|
Galaj E, Bi GH, Xi ZX. β-caryophyllene inhibits heroin self-administration, but does not alter opioid-induced antinociception in rodents. Neuropharmacology 2024; 252:109947. [PMID: 38631564 DOI: 10.1016/j.neuropharm.2024.109947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
A growing body of research indicates that β-caryophyllene (BCP), a constituent present in a large number of plants, possesses significant therapeutic properties against CNS disorders, including alcohol and psychostimulant use disorders. However, it is unknown whether BCP has similar therapeutic potential for opioid use disorders. In this study, we found that systemic administration of BCP dose-dependently reduced heroin self-administration in rats under an FR2 schedule of reinforcement and partially blocked heroin-enhanced brain stimulation reward in DAT-cre mice, maintained by optical stimulation of midbrain dopamine neurons at high frequencies. Acute administration of BCP failed to block heroin conditioned place preference (CPP) in male mice, but attenuated heroin-induced CPP in females. Furthermore, repeated dosing with BCP for 5 days facilitated the extinction of CPP in female but not male mice. In the hot plate assay, pretreatment with the same doses of BCP failed to enhance or prolong opioid antinociception. Lastly, in a substitution test, BCP replacement for heroin failed to maintain intravenous BCP self-administration, suggesting that BCP itself has no reinforcing properties. These findings suggest that BCP may have certain therapeutic effects against opioid use disorders with fewer unwanted side-effects by itself.
Collapse
Affiliation(s)
- Ewa Galaj
- Department of Psychological and Brain Sciences, Colgate University, Hamilton, NY, USA.
| | - Guo-Hua Bi
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Zheng-Xiong Xi
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
5
|
Badshah I, Anwar M, Murtaza B, Khan MI. Molecular mechanisms of morphine tolerance and dependence; novel insights and future perspectives. Mol Cell Biochem 2024; 479:1457-1485. [PMID: 37470850 DOI: 10.1007/s11010-023-04810-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/06/2023] [Indexed: 07/21/2023]
Abstract
Drug addiction is a devastating condition that poses a serious burden on the society. The use of some drugs like morphine for their tremendous analgesic properties is also accompanied with developing tolerance, dependence and the withdrawal symptoms. These symptoms are frequently severe enough to reinforce the person in recovery to start over the use of drug again and hinder the clinical use of drugs like morphine for chronic pain. Research into opioid receptors and related molecular pathways has seen resurgence in the wake of the growing opioid epidemic. The current study provides a comprehensive scientific exploration of the molecular mechanisms and underlying signalling in morphine tolerance and dependence. It also critically evaluates current therapeutic approaches, shedding light on their efficacy and limitations, and future prospects.
Collapse
Affiliation(s)
- Ismail Badshah
- Riphah Institute of Pharmaceutical Sciences, G-7/4 Campus, Islamabad, Pakistan
| | - Maira Anwar
- Riphah Institute of Pharmaceutical Sciences, G-7/4 Campus, Islamabad, Pakistan
| | - Babar Murtaza
- Riphah Institute of Pharmaceutical Sciences, G-7/4 Campus, Islamabad, Pakistan.
| | - Muhammad Imran Khan
- Department of Biomedical Sciences, Pak Austria Fachhochschule: Institute of Applied Sciences and Technology, Haripur, Khyber Pakhtunkhwa, Pakistan.
| |
Collapse
|
6
|
Morosini C, Vivarelli F, Rullo L, Volino E, Losapio LM, Paolini M, Romualdi P, Canistro D, Candeletti S. Unburned Tobacco Smoke Affects Neuroinflammation-Related Pathways in the Rat Mesolimbic System. Int J Mol Sci 2024; 25:5259. [PMID: 38791298 PMCID: PMC11120663 DOI: 10.3390/ijms25105259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Tobacco use disorder represents a significant public health challenge due to its association with various diseases. Despite awareness efforts, smoking rates remain high, partly due to ineffective cessation methods and the spread of new electronic devices. This study investigated the impact of prolonged nicotine exposure via a heat-not-burn (HnB) device on selected genes and signaling proteins involved in inflammatory processes in the rat ventral tegmental area (VTA) and nucleus accumbens (NAc), two brain regions associated with addiction to different drugs, including nicotine. The results showed a reduction in mRNA levels for PPARα and PPARγ, two nuclear receptors and anti-inflammatory transcription factors, along with the dysregulation of gene expression of the epigenetic modulator KDM6s, in both investigated brain areas. Moreover, decreased PTEN mRNA levels and higher AKT phosphorylation were detected in the VTA of HnB-exposed rats with respect to their control counterparts. Finally, significant alterations in ERK 1/2 phosphorylation were observed in both mesolimbic areas, with VTA decrease and NAc increase, respectively. Overall, the results suggest that HnB aerosol exposure disrupts intracellular pathways potentially involved in the development and maintenance of the neuroinflammatory state. Moreover, these data highlight that, similar to conventional cigarettes, HnB devices use affects specific signaling pathways shaping neuroinflammatory process in the VTA and NAc, thus triggering mechanisms that are currently considered as potentially relevant for the development of addictive behavior.
Collapse
Affiliation(s)
- Camilla Morosini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum–University of Bologna, 40126 Bologna, Italy; (C.M.); (F.V.); (E.V.); (L.M.L.); (M.P.); (D.C.); (S.C.)
| | - Fabio Vivarelli
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum–University of Bologna, 40126 Bologna, Italy; (C.M.); (F.V.); (E.V.); (L.M.L.); (M.P.); (D.C.); (S.C.)
| | - Laura Rullo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum–University of Bologna, 40126 Bologna, Italy; (C.M.); (F.V.); (E.V.); (L.M.L.); (M.P.); (D.C.); (S.C.)
| | - Emilia Volino
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum–University of Bologna, 40126 Bologna, Italy; (C.M.); (F.V.); (E.V.); (L.M.L.); (M.P.); (D.C.); (S.C.)
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum–University of Bologna, 40126 Bologna, Italy
| | - Loredana Maria Losapio
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum–University of Bologna, 40126 Bologna, Italy; (C.M.); (F.V.); (E.V.); (L.M.L.); (M.P.); (D.C.); (S.C.)
| | - Moreno Paolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum–University of Bologna, 40126 Bologna, Italy; (C.M.); (F.V.); (E.V.); (L.M.L.); (M.P.); (D.C.); (S.C.)
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum–University of Bologna, 40126 Bologna, Italy; (C.M.); (F.V.); (E.V.); (L.M.L.); (M.P.); (D.C.); (S.C.)
| | - Donatella Canistro
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum–University of Bologna, 40126 Bologna, Italy; (C.M.); (F.V.); (E.V.); (L.M.L.); (M.P.); (D.C.); (S.C.)
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum–University of Bologna, 40126 Bologna, Italy; (C.M.); (F.V.); (E.V.); (L.M.L.); (M.P.); (D.C.); (S.C.)
| |
Collapse
|
7
|
Grodin EN. Neuroimmune modulators as novel pharmacotherapies for substance use disorders. Brain Behav Immun Health 2024; 36:100744. [PMID: 38435721 PMCID: PMC10906159 DOI: 10.1016/j.bbih.2024.100744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/20/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
One promising avenue of research is the use of neuroimmune modulators to treat substance use disorders (SUDs). Neuroimmune modulators target the interactions between the nervous system and immune system, which have been found to play a crucial role in the development and maintenance of SUDs. Multiple classes of substances produce alterations to neuroimmune signaling and peripheral immune function, including alcohol, opioids, and psychostimulants Preclinical studies have shown that neuroimmune modulators can reduce drug-seeking behavior and prevent relapse in animal models of SUDs. Additionally, early-phase clinical trials have demonstrated the safety and feasibility of using neuroimmune modulators as a treatment for SUDs in humans. These therapeutics can be used as stand-alone treatments or as adjunctive. This review summarizes the current state of the field and provides future directions with a specific focus on personalized medicine.
Collapse
Affiliation(s)
- Erica N. Grodin
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
- Cousins Center for Psychoneuroimmunology, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
8
|
Doyle MR, Martinez AR, Qiao R, Dirik S, Di Ottavio F, Pascasio G, Martin-Fardon R, Benner C, George O, Telese F, de Guglielmo G. Strain and sex-related behavioral variability of oxycodone dependence in rats. Neuropharmacology 2023; 237:109635. [PMID: 37327971 PMCID: PMC10353778 DOI: 10.1016/j.neuropharm.2023.109635] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/18/2023]
Abstract
Over the past two decades, the escalating prescription of opioid medications for pain management has culminated in a widespread opioid epidemic, significantly impacting public health, social dynamics, and economic stability. The urgent need for improved treatments for opioid addiction necessitates a deeper understanding of its biological underpinnings, with genetic variations playing a crucial role in individual susceptibility to opioid use disorder (OUD) and influencing clinical practices. In this study, we leverage the genetic diversity of four rat strains (ACI/N, BN/NHsd, WKY/N, and F344/N) to examine the contribution of genetic factors to oxycodone metabolism and addiction-like behaviors. We used the extended access to intravenous oxycodone self-administration procedure (12 h/day, 0.15 mg/kg/injection) to comprehensively characterize oxycodone-related behaviors and pharmacokinetics. We measured escalation of oxycodone self-administration, motivation for drug consumption, tolerance to the analgesic effects of oxycodone, withdrawal-induced hyperalgesia, and oxycodone-induced respiratory depression. Additionally, we examined oxycodone-seeking behavior after four weeks of withdrawal by reintroducing the animals to environmental and cue stimuli previously associated with oxycodone self-administration. The findings revealed notable strain differences in several behavioral measures, including oxycodone metabolism. Intriguingly, BN/NHsd and WKY/N strains exhibited similar drug intake and escalation patterns but displayed significant disparities in oxycodone and oxymorphone metabolism. Minimal sex differences were observed within strains, primarily relating to oxycodone metabolism. In conclusion, this study identifies strain differences in the behavioral responses and pharmacokinetics associated with oxycodone self-administration in rats, providing a robust foundation for identifying genetic and molecular variants associated with various facets of the opioid addiction process.
Collapse
Affiliation(s)
- Michelle R Doyle
- Department of Psychiatry, University of California San Diego, School of Medicine, La Jolla, CA, USA; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Angelica R Martinez
- Department of Psychiatry, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Ran Qiao
- Department of Psychiatry, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Selen Dirik
- Department of Psychiatry, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Francesca Di Ottavio
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Glenn Pascasio
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Rémi Martin-Fardon
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Christopher Benner
- Department of Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Olivier George
- Department of Psychiatry, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Francesca Telese
- Department of Psychiatry, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Giordano de Guglielmo
- Department of Psychiatry, University of California San Diego, School of Medicine, La Jolla, CA, USA.
| |
Collapse
|
9
|
Butelman ER, Goldstein RZ, Nwaneshiudu CA, Girdhar K, Roussos P, Russo SJ, Alia-Klein N. Neuroimmune Mechanisms of Opioid Use Disorder and Recovery: Translatability to Human Studies, and Future Research Directions. Neuroscience 2023; 528:102-116. [PMID: 37562536 PMCID: PMC10720374 DOI: 10.1016/j.neuroscience.2023.07.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023]
Abstract
Opioid use disorder (OUD) is a major current cause of morbidity and mortality. Long-term exposure to short-acting opioids (MOP-r agonists such as heroin or fentanyl) results in complex pathophysiological changes to neuroimmune and neuroinflammatory functions, affected in part by peripheral mechanisms (e.g., cytokines in blood), and by neuroendocrine systems such as the hypothalamic-pituitary-adrenal (HPA) stress axis. There are important findings from preclinical models, but their role in the trajectory and outcomes of OUD in humans is not well understood. The goal of this narrative review is to examine available data on immune and inflammatory functions in persons with OUD, and to identify major areas for future research. Peripheral blood biomarker studies revealed a pro-inflammatory state in persons with OUD in withdrawal or early abstinence, consistent with available postmortem brain studies (which show glial activation) and diffusion tensor imaging studies (indicating white matter disruptions), with gradual abstinence-associated recovery. The mechanistic roles of these neuroimmune and neuroinflammatory changes in the trajectory of OUD (including recovery and medication management) cannot be examined practically with postmortem data. Collection of longitudinal data in larger-scale human cohorts would allow examination of these mechanisms associated with OUD stage and progression. Given the heterogeneity in presentation of OUD, a precision medicine approach integrating multi-omic peripheral biomarkers and comprehensive phenotyping, including neuroimaging, can be beneficial in risk stratification, and individually optimized selection of interventions for individuals who will benefit, and assessments under refractory therapy.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Depts. of Psychiatry and Neuroscience, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Rita Z Goldstein
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Depts. of Psychiatry and Neuroscience, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chinwe A Nwaneshiudu
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiran Girdhar
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA, Medical Center, Bronx, NY, USA
| | - Scott J Russo
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Brain and Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nelly Alia-Klein
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Depts. of Psychiatry and Neuroscience, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
10
|
Lunerti V, Shen Q, Li H, Benvenuti F, Soverchia L, Narendran R, Weiss F, Cannella N, Ciccocioppo R. Cebranopadol, a novel long-acting opioid agonist with low abuse liability, to treat opioid use disorder: Preclinical evidence of efficacy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.21.550008. [PMID: 37546836 PMCID: PMC10401954 DOI: 10.1101/2023.07.21.550008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The gold standard pharmacological treatment for opioid use disorder (OUD) consists of maintenance therapy with long-acting opioid agonists such as buprenorphine and methadone. Despite these compounds having demonstrated substantial efficacy, a significant number of patients do not show optimal therapeutic responses. Moreover, the abuse liability of these medications remains a major concern. Cebranopadol, is a new, long-acting pan-opioid agonist that also activates the nociception/orphanin FQ NOP receptor. Here we used rats to explore the therapeutic potential of this agent in OUD. First, in operant intravenous self-administration experiments we compared the potential abuse liability of cebranopadol with the prototypical opioid heroin. Under a fixed ratio 1 (FR1) contingency, rats maintained responding for heroin (1, 7, 20, 60 μg/inf) to a larger extent than cebranopadol (0.03, 0.1, 0.3, 1.0, 6.0 μg/inf). When the contingency was switched to a progressive ratio (PR) reinforcement schedule, heroin maintained responding at high levels at all except the lowest dose. Conversely, in the cebranopadol groups responding decreased drastically and the break point (BP) did not differ from saline controls. Next, we demonstrated that oral administration of cebranopadol (0, 25, 50 μg/kg) significantly attenuated drug self-administration independent of heroin dose (1, 7, 20, 60 μg/inf). Cebranopadol also reduced the break point for heroin (20 μg/inf). Furthermore, in a heroin self-administration training extinction/reinstatement paradigm, pretreatment with cebranopadol significantly attenuated yohimbine stress-induced reinstatement of drug seeking. Together, these data indicate that cebranopadol has limited abuse liability compared to heroin and is highly efficacious in attenuating opioid self-administration and stress-induced reinstatement, suggesting clinical potential of this compound for OUD treatment.
Collapse
|
11
|
Monroe SC, Radke AK. Opioid withdrawal: role in addiction and neural mechanisms. Psychopharmacology (Berl) 2023; 240:1417-1433. [PMID: 37162529 PMCID: PMC11166123 DOI: 10.1007/s00213-023-06370-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/19/2023] [Indexed: 05/11/2023]
Abstract
Withdrawal from opioids involves a negative affective state that promotes maintenance of drug-seeking behavior and relapse. As such, understanding the neurobiological mechanisms underlying withdrawal from opioid drugs is critical as scientists and clinicians seek to develop new treatments and therapies. In this review, we focus on the neural systems known to mediate the affective and somatic signs and symptoms of opioid withdrawal, including the mesolimbic dopaminergic system, basolateral amygdala, extended amygdala, and brain and hormonal stress systems. Evidence from preclinical studies suggests that these systems are altered following opioid exposure and that these changes mediate behavioral signs of negative affect such as aversion and anxiety during withdrawal. Adaptations in these systems also parallel the behavioral and psychological features of opioid use disorder (OUD), highlighting the important role of withdrawal in the development of addictive behavior. Implications for relapse and treatment are discussed as well as promising avenues for future research, with the hope of promoting continued progress toward characterizing neural contributors to opioid withdrawal and compulsive opioid use.
Collapse
Affiliation(s)
- Sean C Monroe
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, 90 N Patterson Ave, Oxford, OH, USA
| | - Anna K Radke
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, 90 N Patterson Ave, Oxford, OH, USA.
| |
Collapse
|
12
|
Kuhn BN, Cannella N, Crow AD, Roberts AT, Lunerti V, Allen C, Nall RW, Hardiman G, Woods LCS, Chung D, Ciccocioppo R, Kalivas PW. Novelty-induced locomotor behavior predicts heroin addiction vulnerability in male, but not female, rats. Psychopharmacology (Berl) 2022; 239:3605-3620. [PMID: 36112154 PMCID: PMC9632364 DOI: 10.1007/s00213-022-06235-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/02/2022] [Indexed: 11/28/2022]
Abstract
RATIONALE The ongoing rise in opioid use disorder (OUD) has made it imperative to better model the individual variation within the human population that contributes to OUD vulnerability. Using animal models that capture such variation can be a useful tool. Individual variation in novelty-induced locomotion is predictive of substance use disorder (SUD) propensity. In this model, rats are characterized as high-responders (HR) or low-responders (LR) using a median split based on distance travelled during a locomotor test, and HR rats are generally found to exhibit a more SUD vulnerable behavioral phenotype. OBJECTIVES The HR/LR model has commonly been used to assess behaviors in male rats using psychostimulants, with limited knowledge of the predictive efficacy of this model in females or the use of an opioid as the reward. In the current study, we assessed several behaviors across the different phases of drug addiction (heroin taking, refraining, and seeking) in over 500 male and female heterogeneous stock rats run at two geographically separate locations. Rats were characterized as HRs or LRs within each sex for analysis. RESULTS Overall, females exhibit a more OUD vulnerable phenotype relative to males. Additionally, the HR/LR model was predictive of OUD-like behaviors in male, but not female rats. Furthermore, phenotypes did not differ in anxiety-related behaviors, reacquisition of heroin-taking, or punished heroin-taking behavior in either sex. CONCLUSIONS These results emphasize the importance of assessing females in models of individual variation in SUD and highlight limitations in using the HR/LR model to assess OUD propensity.
Collapse
Affiliation(s)
- Brittany N Kuhn
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC, 29425, USA.
| | | | - Ayteria D Crow
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC, 29425, USA
| | - Analyse T Roberts
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC, 29425, USA
| | | | - Carter Allen
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Rusty W Nall
- Department of Psychology, Jacksonville State University, Jacksonville, AL, USA
| | - Gary Hardiman
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | | | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | | | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC, 29425, USA
| |
Collapse
|
13
|
Gipson CD, Dunn KE, Bull A, Ulangkaya H, Hossain A. Establishing preclinical withdrawal syndrome symptomatology following heroin self-administration in male and female rats. Exp Clin Psychopharmacol 2021; 29:636-649. [PMID: 32297787 PMCID: PMC8405057 DOI: 10.1037/pha0000375] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Opioid use disorder (OUD) is a significant health problem, and understanding mechanisms of various aspects of OUD including drug use and withdrawal is important. Preclinical models provide an ideal opportunity to evaluate mechanisms underlying opioid withdrawal. Current models are limited by their reliance upon forced opioid administration, focus on the acute (and not protracted) syndrome, and exclusion of females. In this study, male and female rats self-administered heroin (maintenance dose of 12.5 μg/kg/infusion) and opioid withdrawal after abrupt discontinuation was measured. In Phase 1, acute withdrawal symptoms were rated in male and female rats at 0, 16, 48, and 72 hr after the last self-administration session. Total somatic signs increased until 48 hr (predominantly in females), and heroin intake positively correlated with total somatic signs at the 48 and 72 hr timepoints. Measures of hyperactivity and anxiety-like behavior increased by 16 and 48 hr, respectively. In Phase 2, symptoms were assessed at baseline, acute, and protracted (168 and 312 hr after self-administration) timepoints in a subset of male and female rats from Phase 1. The total number of somatic signs did not differ across timepoints, though females displayed significantly higher body temperature at all timepoints compared with males, indicating sex-specific protracted withdrawal symptomatology. These data provide a thorough characterization of rodent opioid withdrawal symptomatology after self-administration and abrupt discontinuation that serve as a foundation for future studies designed to mimic the human experience, and demonstrate the importance of characterizing acute and protracted withdrawal with sex-specificity in preclinical models of opioid self-administration. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
Affiliation(s)
- Cassandra D. Gipson
- Department of Family and Community Medicine, University of Kentucky, Lexington, KY
| | - Kelly E. Dunn
- Behavioral Pharmacology Research Unit, Johns Hopkins University, Baltimore, MD
| | - Amanda Bull
- Department of Psychology, Arizona State University, Tempe, AZ
| | - Hanaa Ulangkaya
- Department of Psychology, Arizona State University, Tempe, AZ
| | - Aronee Hossain
- Department of Psychology, Arizona State University, Tempe, AZ
| |
Collapse
|
14
|
Rahman S, Rahman ZI, Ronan PJ, Lutfy K, Bell RL. Adolescent opioid abuse: Role of glial and neuroimmune mechanisms. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:147-165. [PMID: 34801168 DOI: 10.1016/bs.irn.2021.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Opioids are widely prescribed for pain management, and prescription opioid misuse in adolescents has become a major epidemic in the United States and worldwide. Emerging data indicate that adolescence represents a critical period of brain development, and exposure to opioids during adolescence may increase the risk of addiction in adulthood. There is growing evidence that disruptions in brain glial function may be implicated in numerous chronic neuropathologies. Evidence suggests that glial mechanisms have an important role in the development and maintenance of opioid abuse and the risk for addiction. This review will describe glial and neuroimmune mechanisms involved in opioid use disorders during adolescence, which may increase substance use disorder liability later in life. Moreover, this review will identify some important neuro-glial targets, involved in opioid abuse and addiction, to develop future preventions and treatment strategies.
Collapse
Affiliation(s)
- S Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, United States.
| | - Z I Rahman
- University of Minnesota Medical School, Minneapolis, MN, United States
| | - P J Ronan
- Department of Psychiatry and Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, United States; Research Service, Sioux Falls VA Healthcare System, Sioux Falls, SD, United States
| | - K Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - R L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
15
|
Galaj E, Bi GH, Moore A, Chen K, He Y, Gardner E, Xi ZX. Beta-caryophyllene inhibits cocaine addiction-related behavior by activation of PPARα and PPARγ: repurposing a FDA-approved food additive for cocaine use disorder. Neuropsychopharmacology 2021; 46:860-870. [PMID: 33069159 PMCID: PMC8026612 DOI: 10.1038/s41386-020-00885-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/15/2020] [Accepted: 10/05/2020] [Indexed: 01/09/2023]
Abstract
Cocaine abuse continues to be a serious health problem worldwide. Despite intense research, there is still no FDA-approved medication to treat cocaine use disorder (CUD). In this report, we explored the potential utility of beta-caryophyllene (BCP), an FDA-approved food additive for the treatment of CUD. We found that BCP, when administered intraperitoneally or intragastrically, dose-dependently attenuated cocaine self-administration, cocaine-conditioned place preference, and cocaine-primed reinstatement of drug seeking in rats. In contrast, BCP failed to alter food self-administration or cocaine-induced hyperactivity. It also failed to maintain self-administration in a drug substitution test, suggesting that BCP has no abuse potential. BCP was previously reported to be a selective CB2 receptor agonist. Unexpectedly, pharmacological blockade or genetic deletion of CB1, CB2, or GPR55 receptors in gene-knockout mice failed to alter BCP's action against cocaine self-administration, suggesting the involvement of non-CB1, non-CB2, and non-GPR55 receptor mechanisms. Furthermore, pharmacological blockade of μ opioid receptor or Toll-like receptors complex failed to alter, while blockade of peroxisome proliferator-activated receptors (PPARα, PPARγ) reversed BCP-induced reduction in cocaine self-administration, suggesting the involvement of PPARα and PPARγ in BCP's action. Finally, we used electrical and optogenetic intracranial self-stimulation (eICSS, oICSS) paradigms to study the underlying neural substrate mechanisms. We found that BCP is more effective in attenuation of cocaine-enhanced oICSS than eICSS, the former driven by optical activation of midbrain dopamine neurons in DAT-cre mice. These findings indicate that BCP may be useful for the treatment of CUD, likely by stimulation of PPARα and PPARγ in the mesolimbic system.
Collapse
Affiliation(s)
- Ewa Galaj
- grid.420090.f0000 0004 0533 7147Addiction Biology Unit, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224 USA
| | - Guo-Hua Bi
- grid.420090.f0000 0004 0533 7147Addiction Biology Unit, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224 USA
| | - Allamar Moore
- grid.420090.f0000 0004 0533 7147Neuropychopharmacology Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224 USA
| | - Kai Chen
- grid.420090.f0000 0004 0533 7147Addiction Biology Unit, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224 USA ,grid.413247.7Present Address: Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071 China
| | - Yi He
- grid.420090.f0000 0004 0533 7147Addiction Biology Unit, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224 USA ,grid.21925.3d0000 0004 1936 9000Present Address: Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Eliot Gardner
- grid.420090.f0000 0004 0533 7147Neuropychopharmacology Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224 USA
| | - Zheng-Xiong Xi
- Addiction Biology Unit, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA.
| |
Collapse
|
16
|
Stopponi S, Fotio Y, Cifani C, Li H, Haass-Koffler CL, Cannella N, Demopulos G, Gaitanaris G, Ciccocioppo R. Andrographis paniculata and Its Main Bioactive Ingredient Andrographolide Decrease Alcohol Drinking and Seeking in Rats Through Activation of Nuclear PPARγ Pathway. Alcohol Alcohol 2021; 56:240-249. [PMID: 33401299 DOI: 10.1093/alcalc/agaa136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND AIMS Andrographis paniculata is an annual herbaceous plant which belongs to the Acanthaceae family. Extracts from this plant have shown hepatoprotective, anti-inflammatory and antidiabetic properties, at least in part, through activation of the nuclear receptor Peroxisome Proliferator-Activated Receptor-gamma (PPAR γ). Recent evidence has demonstrated that activation of PPARγ reduces alcohol drinking and seeking in Marchigian Sardinian (msP) alcohol-preferring rats. METHODS The present study evaluated whether A. paniculata reduces alcohol drinking and relapse in msP rats by activating PPARγ. RESULTS Oral administration of an A. paniculata dried extract (0, 15, 150 mg/kg) lowered voluntary alcohol consumption in a dose-dependent manner and achieved ~65% reduction at the dose of 450 mg/kg. Water and food consumption were not affected by the treatment. Administration of Andrographolide (5 and 10 mg/kg), the main active component of A. paniculata, also reduced alcohol drinking. This effect was suppressed by the selective PPARγ antagonist GW9662. Subsequently, we showed that oral administration of A. paniculata (0, 150, 450 mg/kg) prevented yohimbine- but not cues-induced reinstatement of alcohol seeking. CONCLUSIONS Results point to A. paniculata-mediated PPARγactivation as a possible therapeutic strategy to treat alcohol use disorder.
Collapse
Affiliation(s)
- Serena Stopponi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Yannick Fotio
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy.,Department of Anatomy and Neurobiology, School of Medicine, University of California, 807 Health Science Road, 92617 Irvine, USA
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Hongwu Li
- College of Chemical Engineering, Changchun University of Technology, 2055 Yan An Road, Chao Yang District, 130021 Changchun, China
| | - Carolina L Haass-Koffler
- Center Alcohol and Addiction Studies, Department Psychiatry and Human Behavior Department Behavioral and Social Sciences Brow University 121 S. Main Street, Providence, RI 02931, USA
| | - Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Gregory Demopulos
- Center Alcohol and Addiction Studies, Department Psychiatry and Human Behavior Department Behavioral and Social Sciences Brow University 121 S. Main Street, Providence, RI 02931, USA
| | - George Gaitanaris
- Center Alcohol and Addiction Studies, Department Psychiatry and Human Behavior Department Behavioral and Social Sciences Brow University 121 S. Main Street, Providence, RI 02931, USA.,Omeros Corporation, 201 Elliot Avenue West, Seattle, WA 98119, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| |
Collapse
|
17
|
Vanaveski T, Molchanova S, Pham DD, Schäfer A, Pajanoja C, Narvik J, Srinivasan V, Urb M, Koivisto M, Vasar E, Timmusk T, Minkeviciene R, Eriksson O, Lalowski M, Taira T, Korhonen L, Voikar V, Lindholm D. PGC-1α Signaling Increases GABA(A) Receptor Subunit α2 Expression, GABAergic Neurotransmission and Anxiety-Like Behavior in Mice. Front Mol Neurosci 2021; 14:588230. [PMID: 33597848 PMCID: PMC7882546 DOI: 10.3389/fnmol.2021.588230] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/14/2021] [Indexed: 12/23/2022] Open
Abstract
Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is a master regulator of mitochondria biogenesis and cell stress playing a role in metabolic and degenerative diseases. In the brain PGC-1α expression has been localized mainly to GABAergic interneurons but its overall role is not fully understood. We observed here that the protein levels of γ-aminobutyric acid (GABA) type A receptor-α2 subunit (GABARα2) were increased in hippocampus and brain cortex in transgenic (Tg) mice overexpressing PGC-1α in neurons. Along with this, GABARα2 expression was enhanced in the hippocampus of the PGC-1α Tg mice, as shown by quantitative PCR. Double immunostaining revealed that GABARα2 co-localized with the synaptic protein gephyrin in higher amounts in the striatum radiatum layer of the hippocampal CA1 region in the Tg compared with Wt mice. Electrophysiology revealed that the frequency of spontaneous and miniature inhibitory postsynaptic currents (mIPSCs) was increased in the CA1 region in the Tg mice, indicative of an augmented GABAergic transmission. Behavioral tests revealed an increase for anxiety-like behavior in the PGC-1α Tg mice compared with controls. To study whether drugs acting on PPARγ can affect GABARα2, we employed pioglitazone that elevated GABARα2 expression in primary cultured neurons. Similar results were obtained using the specific PPARγ agonist, N-(2-benzoylphenyl)-O-[2-(methyl-2-pyridinylamino) ethyl]-L-tyrosine hydrate (GW1929). These results demonstrate that PGC-1α regulates GABARα2 subunits and GABAergic neurotransmission in the hippocampus with behavioral consequences. This indicates further that drugs like pioglitazone, widely used in the treatment of type 2 diabetes, can influence GABARα2 expression via the PPARγ/PGC-1α system.
Collapse
Affiliation(s)
- Taavi Vanaveski
- Medicum, Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Helsinki, Finland.,Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Quretec Ltd., Tartu, Estonia
| | - Svetlana Molchanova
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Dan Duc Pham
- Medicum, Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Annika Schäfer
- Medicum, Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Ceren Pajanoja
- Medicum, Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Jane Narvik
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Quretec Ltd., Tartu, Estonia
| | - Vignesh Srinivasan
- Medicum, Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | | | - Maria Koivisto
- Medicum, Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Eero Vasar
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Tönis Timmusk
- Protobios LCC, Tallinn, Estonia.,Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | | | - Ove Eriksson
- Medicum, Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Maciej Lalowski
- Medicum, Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.,Meilahti Clinical Proteomics Core Facility, HiLIFE, University of Helsinki, Helsinki, Finland.,Department of Biomedical Proteomics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznań, Poland
| | - Tomi Taira
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine and Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Laura Korhonen
- Department of Child and Adolescent Psychiatry and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Vootele Voikar
- Neuroscience Center and Laboratory Animal Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Dan Lindholm
- Medicum, Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Helsinki, Finland
| |
Collapse
|
18
|
Verster JC, Scholey A, Dahl TA, Iversen JM. Functional observation after morphine withdrawal: effects of SJP-005. Psychopharmacology (Berl) 2021; 238:1449-1460. [PMID: 33555386 PMCID: PMC8139893 DOI: 10.1007/s00213-021-05771-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 01/21/2021] [Indexed: 10/27/2022]
Abstract
RATIONALE AND OBJECTIVE SJP-005 (ketotifen and ibuprofen) is being developed as a potential new treatment for opioid withdrawal. Three studies were conducted to evaluate the early phase (acute, day 1) and late phase (days 2-12) effects of SJP-005 on discontinuation-induced morphine withdrawal. METHODS Sprague-Dawley rats received subcutaneous morphine twice daily for 18 days and ceased on day 19. Twice daily, oral dosages of placebo or SJP-005 (1 mg/kg ketotifen and 15 mg/kg ibuprofen) were administered starting 4 days before (study 1), 2 days before (study 2), or immediately after (study 3) morphine cessation. Functional observations were made up to 12 h after treatment cessation on day 19 (early phase), and immediately after treatment on days 20-30 (late phase). Treatment effects (mean overall score, and individual symptoms) were compared with placebo using ANOVA, and Tukey's tests in case of multiple comparisons. RESULTS Across the studies, the number of withdrawal signs on day 19 (early phase) and days 20-30 (late phase) was lower with SJP-005 compared with placebo. The effects of SJP-005 when treatment was initiated 2 days before morphine cessation by discontinuation were most pronounced and statistically significant in the late phase (F(1,18) = 14.10, p = 0.001). In particular, a significant reduction was observed in hypersensitivity to touch (F(1,18) = 13.65, p = 0.002). A 50% reduction in withdrawal symptoms was observed 9.0 days after placebo versus 4.5 days after SJP-005. After 9.0 days, all withdrawal symptoms were absent in the SJP-005 group, while symptoms in the placebo group were still evident on day 18. CONCLUSION Compared to placebo, SJP-005 significantly reduced the incidence and duration of discontinuation-induced morphine withdrawal symptoms when treatment was initiated 2 days before morphine cessation.
Collapse
Affiliation(s)
- Joris C. Verster
- grid.5477.10000000120346234Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584CG, Utrecht, The Netherlands ,grid.1027.40000 0004 0409 2862Centre for Human Psychopharmacology, Swinburne University, Melbourne, VIC 3122 Australia ,grid.5477.10000000120346234Psychopharmacology, Utrecht University, Po Box 80082, 3508TB Utrecht, Netherlands
| | - Andrew Scholey
- grid.1027.40000 0004 0409 2862Centre for Human Psychopharmacology, Swinburne University, Melbourne, VIC 3122 Australia
| | - Thomas A. Dahl
- Sen-Jam Pharmaceutical, 223 Wall St., #130, Huntington, NY 11743 USA
| | | |
Collapse
|
19
|
Ciccocioppo R, Ubaldi M. Nuclear peroxisome proliferator activated receptor-gamma (PPARγ) as a therapeutic target to treat neurodegeneration and dependence elicited by drugs of abuse. Neural Regen Res 2021; 16:984-985. [PMID: 33229744 PMCID: PMC8178789 DOI: 10.4103/1673-5374.297072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| |
Collapse
|
20
|
Jung T, Hudson R, Rushlow W, Laviolette SR. Functional interactions between cannabinoids, omega-3 fatty acids, and peroxisome proliferator-activated receptors: Implications for mental health pharmacotherapies. Eur J Neurosci 2020; 55:1088-1100. [PMID: 33108021 DOI: 10.1111/ejn.15023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/08/2020] [Accepted: 10/16/2020] [Indexed: 12/18/2022]
Abstract
Cannabis contains a plethora of phytochemical constituents with diverse neurobiological effects. Cannabidiol (CBD) is the main non-psychotropic component found in cannabis that is capable of modulating mesocorticolimbic DA transmission and may possess therapeutic potential for several neuropsychiatric disorders. Emerging evidence also suggests that, similar to CBD, omega-3 polyunsaturated fatty acids may regulate DA transmission and possess therapeutic potential for similar neuropsychiatric disorders. Although progress has been made to elucidate the mechanisms underlying the therapeutic properties of CBD and omega-3s, it remains unclear through which receptor mechanisms they may produce their purported effects. Peroxisome proliferator-activated receptors are a group of nuclear transcription factors with multiple isoforms. PPARγ is an isoform activated by both CBD and omega-3, whereas the PPARα isoform is activated by omega-3. Interestingly, the activation of PPARγ and PPARα with selective agonists has been shown to decrease mesocorticolimbic DA activity and block neuropsychiatric symptoms similar to CBD and omega-3s, raising the possibility that CBD and omega-3s produce their effects through PPAR signaling. This review will examine the relationship between CBD, omega-3s, and PPARs and how they may be implicated in the modulation of mesocorticolimbic DAergic abnormalities and associated neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Tony Jung
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.,Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Roger Hudson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.,Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Walter Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.,Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.,Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.,Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.,Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
21
|
Schwandt ML, Diazgranados N, Umhau JC, Kwako LE, George DT, Heilig M. PPARγ activation by pioglitazone does not suppress cravings for alcohol, and is associated with a risk of myopathy in treatment seeking alcohol dependent patients: a randomized controlled proof of principle study. Psychopharmacology (Berl) 2020; 237:2367-2380. [PMID: 32445052 PMCID: PMC11018293 DOI: 10.1007/s00213-020-05540-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 04/27/2020] [Indexed: 12/12/2022]
Abstract
RATIONALE Proinflammatory processes have been implicated in alcohol addiction, craving, and relapse, while studies in experimental animals have suggested that activation of peroxisome proliferator-activated receptor gamma (PPARγ) inhibits proinflammatory signaling. Accordingly, it is hypothesized that medications with PPARγ activity may have therapeutic potential in alcohol dependence. OBJECTIVES We conducted a double-blind, placebo-controlled mechanistic proof of principle study in alcohol-dependent inpatients to investigate the effect of pioglitazone on alcohol craving. METHODS Participants were treated for withdrawal, if needed, and then randomized to pioglitazone (target dose 45 mg/day) or placebo. Once at target dose, they completed two experimental manipulations: guided imagery, which used personalized auditory scripts to induce alcohol cravings, and a low-dose challenge with i.v. lipopolysaccharide (LPS; 0.8 ng/kg) or placebo, on two separate sessions, in counterbalanced order. Behavioral and endocrine responses as well as CSF levels of proinflammatory cytokines were evaluated. RESULTS The study was prematurely terminated after randomization of 16 subjects, following an independent review that established a high risk of myopathy in the active treatment group. Analysis of those who completed the study indicated that pioglitazone was associated with elevated, rather than suppressed alcohol cravings in response to alcohol-associated stimuli. LPS did not induce cravings for alcohol and thus did not lend itself to evaluating pioglitazone effects; however, pioglitazone increased the neuroendocrine stress response to LPS. CSF levels of IL-6, TNF-α, or MCP-1 were unaffected by pioglitazone treatment. CONCLUSIONS Both safety and efficacy biomarker data suggest that pioglitazone lacks potential as a medication for the treatment of alcohol dependence. CLINICAL TRIAL REGISTRATION NCT01631630.
Collapse
Affiliation(s)
- Melanie L Schwandt
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, 10 Center Drive, CRC 1-5330, Bethesda, MD, 20892, USA.
| | - Nancy Diazgranados
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, 10 Center Drive, CRC 1-5330, Bethesda, MD, 20892, USA
| | - John C Umhau
- Center for Drug Evaluation and Research (CDER), United States Food and Drug Administration, Washington, DC, USA
| | - Laura E Kwako
- Division of Treatment and Recovery Research, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - David T George
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, 10 Center Drive, CRC 1-5330, Bethesda, MD, 20892, USA
| | - Markus Heilig
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| |
Collapse
|
22
|
Abstract
The pervasive and devastating nature of substance use disorders underlies the need for the continued development of novel pharmacotherapies. We now know that glia play a much greater role in neuronal processes than once believed. The various types of glial cells (e.g., astrocytes, microglial, oligodendrocytes) participate in numerous functions that are crucial to healthy central nervous system function. Drugs of abuse have been shown to interact with glia in ways that directly contribute to the pharmacodynamic effects responsible for their abuse potential. Through their effect upon glia, drugs of abuse also alter brain function resulting in behavioral changes associated with substance use disorders. Therefore, drug-induced changes in glia and inflammation within the central nervous system (neuroinflammation) have been investigated to treat various aspects of drug abuse and dependence. This article presents a brief overview of the effects of each of the major classes of addictive drugs on glia. Next, the paper reviews the pre-clinical and clinical studies assessing the effects that glial modulators have on abuse-related behavioral effects, such as pleasure, withdrawal, and motivation. There is a strong body of pre-clinical literature demonstrating the general effectiveness of several glia-modulating drugs in models of reward and relapse. Clinical studies have also yielded promising results, though not as robust. There is still much to disentangle regarding the integration between addictive drugs and glial cells. Improved understanding of the relationship between glia and the pathophysiology of drug abuse should allow for more precise exploration in the development and testing of glial-directed treatments for substance use disorders.
Collapse
Affiliation(s)
- Jermaine D. Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
23
|
Matheson J, Le Foll B. Therapeutic Potential of Peroxisome Proliferator-Activated Receptor (PPAR) Agonists in Substance Use Disorders: A Synthesis of Preclinical and Human Evidence. Cells 2020; 9:cells9051196. [PMID: 32408505 PMCID: PMC7291117 DOI: 10.3390/cells9051196] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022] Open
Abstract
Targeting peroxisome proliferator-activated receptors (PPARs) has received increasing interest as a potential strategy to treat substance use disorders due to the localization of PPARs in addiction-related brain regions and the ability of PPAR ligands to modulate dopamine neurotransmission. Robust evidence from animal models suggests that agonists at both the PPAR-α and PPAR-γ isoforms can reduce both positive and negative reinforcing properties of ethanol, nicotine, opioids, and possibly psychostimulants. A reduction in the voluntary consumption of ethanol following treatment with PPAR agonists seems to be the most consistent finding. However, the human evidence is limited in scope and has so far been less promising. There have been no published human trials of PPAR agonists for treatment of alcohol use disorder, despite the compelling preclinical evidence. Two trials of PPAR-α agonists as potential smoking cessation drugs found no effect on nicotine-related outcomes. The PPAR-γ agonist pioglitazone showed some promise in reducing heroin, nicotine, and cocaine craving in two human laboratory studies and one pilot trial, yet other outcomes were unaffected. Potential explanations for the discordance between the animal and human evidence, such as the potency and selectivity of PPAR ligands and sex-related variability in PPAR physiology, are discussed.
Collapse
Affiliation(s)
- Justin Matheson
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, 27 King’s College Circle, Toronto, ON M5S 3H7, Canada;
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, ON M5S 2S1, Canada
- Correspondence: ; Tel.: +1-416-535-8501 (ext. 34727)
| | - Bernard Le Foll
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, 27 King’s College Circle, Toronto, ON M5S 3H7, Canada;
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, ON M5S 2S1, Canada
- Addictions Division, Centre for Addiction and Mental Health, 100 Stokes Street, Toronto, ON M6J 1H4, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Faculty of Medicine, University of Toronto, 250 College Street, Toronto, ON M5T 1R8, Canada
- Institute of Medical Sciences, University of Toronto, 1 King’s College Circle, Room 2374, Toronto, ON M5S 1A8, Canada
- Department of Family and Community Medicine, University of Toronto, 500 University Avenue, 5th Floor, Toronto, ON M5G 1V7, Canada
| |
Collapse
|
24
|
Abstract
This paper is the fortieth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2017 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, CUNY, 65-30 Kissena Blvd., Flushing, NY, 11367, United States.
| |
Collapse
|
25
|
Activation of GLP-1 receptors attenuates oxycodone taking and seeking without compromising the antinociceptive effects of oxycodone in rats. Neuropsychopharmacology 2020; 45:451-461. [PMID: 31581176 PMCID: PMC6969180 DOI: 10.1038/s41386-019-0531-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022]
Abstract
Despite the effectiveness of current medications to treat opioid use disorder, there is still a high rate of relapse following detoxification. Thus, there is critical need for innovative studies aimed at identifying novel neurobiological mechanisms that could be targeted to treat opioid use disorder. A growing body of preclinical evidence indicates that glucagon-like peptide-1 (GLP-1) receptor agonists reduce drug reinforcement. However, the efficacy of GLP-1 receptor agonists in attenuating opioid-mediated behaviors has not been thoroughly investigated. Using recently established models of opioid-taking and -seeking behaviors, we showed that systemic administration of the GLP-1 receptor agonist exendin-4 reduced oxycodone self-administration and the reinstatement of oxycodone-seeking behavior in rats. We also identified behaviorally selective doses of exendin-4 that reduced opioid-taking and -seeking behaviors and did not produce adverse feeding effects in oxycodone-experienced rats. To identify a central site of action, we showed that systemic exendin-4 penetrated the brain and bound putative GLP-1 receptors on dopamine D1 receptor- and dopamine D2 receptor-expressing medium spiny neurons in the nucleus accumbens shell. Consistent with our systemic studies, infusions of exendin-4 directly into the accumbens shell attenuated oxycodone self-administration and the reinstatement of oxycodone-seeking behavior without affecting ad libitum food intake. Finally, exendin-4 did not alter the analgesic effects of oxycodone, suggesting that activation of GLP-1 receptors attenuated opioid reinforcement without reducing the thermal antinociceptive effects of oxycodone. Taken together, these findings suggest that GLP-1 receptors could serve as potential molecular targets for pharmacotherapies aimed at reducing opioid use disorder.
Collapse
|
26
|
Abstract
This chapter describes recent clinical trials for opioid use disorder (OUD), an area that has rapidly accelerated in response to the opioid overdose crisis in the USA and newly appropriated funding. Trials involve a wide range of compounds including cannabinoids and psychedelics, new and existing compounds targeting domains emerging from addiction neuroscience, agents repurposed from other indications, and novel strategies including vaccines, enzymes, and other biologicals. In parallel, new formulations of existing compounds offer immediate promise, as do a variety of web-based interventions and smartphone-delivered apps. Trials focused on implementing existing effective interventions in mainstream healthcare settings, and others focused on special populations, e.g., adolescents, criminal justice, pregnant women, native Americans, etc., have the potential to vastly expand treatment in the near term. Given the range of ongoing and recent trials, this chapter is not intended to be an exhaustive review but rather to present an overview of approaches within the framework of the opioid treatment cascade and the context of current OUD pharmacotherapies.
Collapse
Affiliation(s)
- Esther Blessing
- Department of Psychiatry, NYU School of Medicine, New York, NY, USA.
| | - Sanya Virani
- Department of Psychiatry, Maimonides Medical Center, Brooklyn, NY, USA
| | - John Rotrosen
- Department of Psychiatry, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
27
|
Glial neuroimmune signaling in opioid reward. Brain Res Bull 2019; 155:102-111. [PMID: 31790721 DOI: 10.1016/j.brainresbull.2019.11.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/20/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Abstract
The opioid epidemic is a growing public concern affecting millions of people worldwide. Opioid-induced reward is the initial and key process leading to opioid abuse and addiction. Therefore, a better understanding of opioid reward may be helpful in developing a treatment for opioid addiction. Emerging evidence suggests that glial cells, particularly microglia and astrocytes, play an essential role in modulating opioid reward. Indeed, glial cells and their associated immune signaling actively regulate neural activity and plasticity, and directly modulate opioid-induced rewarding behaviors. In this review, we describe the neuroimmune mechanisms of how glial cells affect synaptic transmission and plasticity as well as how opioids can activate glial cells affecting the glial-neuronal interaction. Last, we summarize current attempts of applying glial modulators in treating opioid reward.
Collapse
|
28
|
Activation of PPARγ Attenuates the Expression of Physical and Affective Nicotine Withdrawal Symptoms through Mechanisms Involving Amygdala and Hippocampus Neurotransmission. J Neurosci 2019; 39:9864-9875. [PMID: 31685649 DOI: 10.1523/jneurosci.1922-19.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/18/2019] [Indexed: 12/31/2022] Open
Abstract
An isoform of peroxisome proliferator-activated receptors (PPARs), PPARγ, is the receptor for the thiazolidinedione class of anti-diabetic medications including pioglitazone. Neuroanatomical data indicate PPARγ localization in brain areas involved in drug addiction. Preclinical and clinical data have shown that pioglitazone reduces alcohol and opioid self-administration, relapse to drug seeking, and plays a role in emotional responses. Here, we investigated the behavioral effect of PPARγ manipulation on nicotine withdrawal in male Wistar rats and in male mice with neuron-specific PPARγ deletion (PPARγ(-/-)) and their littermate wild-type (PPARγ(+/+)) controls. Real-time quantitative RT-PCR and RNAscope in situ hybridization assays were used for assessing the levels of expression and cell-type localization of PPARγ during nicotine withdrawal. Brain site-specific microinjections of the PPARγ agonist pioglitazone were performed to explore the role of this system on nicotine withdrawal at a neurocircuitry level. Results showed that activation of PPARγ by pioglitazone abolished the expression of somatic and affective nicotine withdrawal signs in rats and in (PPARγ(+/+)) mice. This effect was blocked by the PPARγ antagonist GW9662. During early withdrawal and protracted abstinence, the expression of PPARγ increased in GABAergic and glutamatergic cells of the amygdala and hippocampus, respectively. Hippocampal microinjections of pioglitazone reduced the expression of the physical signs of withdrawal, whereas excessive anxiety associated with protracted abstinence was prevented by pioglitazone microinjection into the amygdala. Our results demonstrate the implication of the neuronal PPARγ in nicotine withdrawal and indicates that activation of PPARγ may offer an interesting strategy for smoking cessation.SIGNIFICANCE STATEMENT Smoking cessation leads the occurrence of physical and affective withdrawal symptoms representing a major burden to quit tobacco use. Here, we show that activation of PPARγ prevents the expression of both somatic and affective signs of nicotine withdrawal. At molecular levels results show that PPARγ expression increases in GABAergic cells in the hippocampus and in GABA- and glutamate-positive cells in the basolateral amygdala. Hippocampal microinjections of pioglitazone reduce the insurgence of the physical withdrawal signs, whereas anxiety linked to protracted abstinence is attenuated by pioglitazone injected into the amygdala. Our results demonstrate the implication of neuronal PPARγ in nicotine withdrawal and suggest that PPARγ agonism may represent a promising treatment to aid smoking cessation.
Collapse
|
29
|
Magrone T, Jirillo E. Drugs of Abuse Induced-Subversion of the Peripheral Immune Response and Central Glial Activity: Focus on Novel Therapeutic Approaches. Endocr Metab Immune Disord Drug Targets 2019; 19:281-291. [PMID: 30488804 DOI: 10.2174/1871530319666181129104329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Drugs of abuse affect both central nervous system (CNS) and peripheral immune function. Besides the involvement of dopamine and glutamate systems, chronic exposure to drugs of abuse alters immune homeostasis, promoting a pro-inflammatory status. At the same time, impaired peripheral immunity leads to an increased susceptibility to infections in drug abusers. DISCUSSION There is evidence that certain drugs, such as opioids, activate microglial cells and astrocytes which, in turn, provoke central neuroinflammation. Particularly, opioids bind the Toll-like receptor (TLR)-4 with increased expression of nuclear factor kappa-light-chain-enhancer of activated B cells and release of pro-inflammatory cytokines. Peripheral mediators released by immune cells also contribute to aggravate central neuroinflammation. CONCLUSION These are based either on the inhibition of TLR-4 activation by drugs of abuse or on the correction of dopamine and glutamate pathways. Finally, a hypothetic nutraceutical intervention with polyphenols in view of their anti-inflammatory and anti-oxidant properties will be outlined as an adjuvant treatment for drugs of abuse-related disorders.
Collapse
Affiliation(s)
- Thea Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, School of Medicine, Bari, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, School of Medicine, Bari, Italy
| |
Collapse
|
30
|
Yammine L, Kosten TR, Pimenova M, Schmitz JM. Cigarette smoking, type 2 diabetes mellitus, and glucagon-like peptide-1 receptor agonists as a potential treatment for smokers with diabetes: An integrative review. Diabetes Res Clin Pract 2019; 149:78-88. [PMID: 30735771 DOI: 10.1016/j.diabres.2019.01.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 01/09/2019] [Accepted: 01/30/2019] [Indexed: 01/26/2023]
Abstract
Tobacco use disorder (TUD), in particular cigarette smoking, contributes significantly to the macro- and micro-vascular complications of type 2 diabetes mellitus (DM). Persons with DM who regularly use tobacco products are twice as likely to experience mortality and negative health outcomes. Despite these risks, TUD remains prevalent in persons with DM. The objective of this integrative review is to summarize the relationship between TUD and DM based on epidemiological and preclinical biological evidence. We conclude with a review of the literature on the glucagon-like peptide-1 (GLP-1) as a potential treatment target for addressing comorbid TUD in smokers with DM.
Collapse
Affiliation(s)
- Luba Yammine
- University of Texas Health Science Center at Houston, Houston, TX, United States.
| | | | - Maria Pimenova
- University of Texas Medical Branch, Galveston, TX, United States
| | - Joy M Schmitz
- University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
31
|
Reiner DJ, Fredriksson I, Lofaro OM, Bossert JM, Shaham Y. Relapse to opioid seeking in rat models: behavior, pharmacology and circuits. Neuropsychopharmacology 2019; 44:465-477. [PMID: 30293087 PMCID: PMC6333846 DOI: 10.1038/s41386-018-0234-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/17/2018] [Accepted: 09/27/2018] [Indexed: 12/13/2022]
Abstract
Lifetime relapse rates remain a major obstacle in addressing the current opioid crisis. Relapse to opioid use can be modeled in rodent studies where drug self-administration is followed by a period of abstinence and a subsequent test for drug seeking. Abstinence can be achieved through extinction training, forced abstinence, or voluntary abstinence. Voluntary abstinence can be accomplished by introducing adverse consequences of continued drug self-administration (e.g., punishment or electric barrier) or by introducing an alternative nondrug reward in a discrete choice procedure (drug versus palatable food or social interaction). In this review, we first discuss pharmacological and circuit mechanisms of opioid seeking, as assessed in the classical extinction-reinstatement model, where reinstatement is induced by reexposure to the self-administered drug (drug priming), discrete cues, discriminative cues, drug-associated contexts, different forms of stress, or withdrawal states. Next, we discuss pharmacological and circuit mechanisms of relapse after forced or voluntary abstinence, including the phenomenon of "incubation of heroin craving" (the time-dependent increases in heroin seeking during abstinence). We conclude by discussing future directions of preclinical relapse-related studies using opioid drugs.
Collapse
Affiliation(s)
- David J. Reiner
- Behavioral Neuroscience Research Branch, IRP-NIDA-NIH, Baltimore, MD USA
| | - Ida Fredriksson
- Behavioral Neuroscience Research Branch, IRP-NIDA-NIH, Baltimore, MD USA
| | - Olivia M. Lofaro
- Behavioral Neuroscience Research Branch, IRP-NIDA-NIH, Baltimore, MD USA
| | | | - Yavin Shaham
- Behavioral Neuroscience Research Branch, IRP-NIDA-NIH, Baltimore, MD, USA.
| |
Collapse
|
32
|
D'Souza MS. Brain and Cognition for Addiction Medicine: From Prevention to Recovery Neural Substrates for Treatment of Psychostimulant-Induced Cognitive Deficits. Front Psychiatry 2019; 10:509. [PMID: 31396113 PMCID: PMC6667748 DOI: 10.3389/fpsyt.2019.00509] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/28/2019] [Indexed: 01/10/2023] Open
Abstract
Addiction to psychostimulants like cocaine, methamphetamine, and nicotine poses a continuing medical and social challenge both in the United States and all over the world. Despite a desire to quit drug use, return to drug use after a period of abstinence is a common problem among individuals dependent on psychostimulants. Recovery for psychostimulant drug-dependent individuals is particularly challenging because psychostimulant drugs induce significant changes in brain regions associated with cognitive functions leading to cognitive deficits. These cognitive deficits include impairments in learning/memory, poor decision making, and impaired control of behavioral output. Importantly, these drug-induced cognitive deficits often impact adherence to addiction treatment programs and predispose abstinent addicts to drug use relapse. Additionally, these cognitive deficits impact effective social and professional rehabilitation of abstinent addicts. The goal of this paper is to review neural substrates based on animal studies that could be pharmacologically targeted to reverse psychostimulant-induced cognitive deficits such as impulsivity and impairment in learning and memory. Further, the review will discuss neural substrates that could be used to facilitate extinction learning and thus reduce emotional and behavioral responses to drug-associated cues. Moreover, the review will discuss some non-pharmacological approaches that could be used either alone or in combination with pharmacological compounds to treat the above-mentioned cognitive deficits. Psychostimulant addiction treatment, which includes treatment for cognitive deficits, will help promote abstinence and allow for better rehabilitation and integration of abstinent individuals into society.
Collapse
Affiliation(s)
- Manoranjan S D'Souza
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, Ada, OH, United States
| |
Collapse
|
33
|
Erickson EK, Grantham EK, Warden AS, Harris RA. Neuroimmune signaling in alcohol use disorder. Pharmacol Biochem Behav 2018; 177:34-60. [PMID: 30590091 DOI: 10.1016/j.pbb.2018.12.007] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/25/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023]
Abstract
Alcohol use disorder (AUD) is a widespread disease with limited treatment options. Targeting the neuroimmune system is a new avenue for developing or repurposing effective pharmacotherapies. Alcohol modulates innate immune signaling in different cell types in the brain by altering gene expression and the molecular pathways that regulate neuroinflammation. Chronic alcohol abuse may cause an imbalance in neuroimmune function, resulting in prolonged perturbations in brain function. Likewise, manipulating the neuroimmune system may change alcohol-related behaviors. Psychiatric disorders that are comorbid with AUD, such as post-traumatic stress disorder, major depressive disorder, and other substance use disorders, may also have underlying neuroimmune mechanisms; current evidence suggests that convergent immune pathways may be involved in AUD and in these comorbid disorders. In this review, we provide an overview of major neuroimmune cell-types and pathways involved in mediating alcohol behaviors, discuss potential mechanisms of alcohol-induced neuroimmune activation, and present recent clinical evidence for candidate immune-related drugs to treat AUD.
Collapse
Affiliation(s)
- Emma K Erickson
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA.
| | - Emily K Grantham
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| | - Anna S Warden
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| | - R A Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| |
Collapse
|
34
|
Evidence of a PPARγ-mediated mechanism in the ability of Withania somnifera to attenuate tolerance to the antinociceptive effects of morphine. Pharmacol Res 2018; 139:422-430. [PMID: 30503841 DOI: 10.1016/j.phrs.2018.11.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/27/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Notwithstanding the experimental evidence indicating Withania somnifera Dunal roots extract (WSE) ability to prolong morphine-elicited analgesia, the mechanisms underlying this effect are largely unknown. With the aim of evaluating a PPARγ-mediated mechanism in such WSE effects, we verified the ability of the PPARγ antagonist GW-9662 to modulate WSE actions. Further, we evaluated the influence of GW-9662 upon WSE / morphine interaction in SH-SY5Y cells since we previously reported that WSE hampers the morphine-induced μ-opioid receptor (MOP) receptor down-regulation. Nociceptive thresholds / tolerance development were assessed in different groups of rats receiving vehicles (control), morphine (10 mg/kg; i.p.), WSE (100 mg/kg, i.p.) and PPARγ antagonist GW-9662 (1 mg/kg; s.c.) in acute and chronic schedules of administration. Moreover, the effects of GW-9662 (5 and 10 μM) applied alone and in combination with morphine (10 μM) and/or WSE (0.25 and 1.00 mg/mL) on the MOP gene expression were investigated in cell cultures. Data analysis revealed a functional effect of the PPARγ antagonist in attenuating the ability of WSE to prolong morphine analgesic effect and to reduce tolerance development after repeated administration. In addition, molecular experiments demonstrated that the blockade of PPARγ by GW-9662 promotes MOP mRNA down-regulation and counteracts the ability of 1.00 mg/mL of WSE to keep an adequate MOP receptor availability. In conclusion, our results support the involvement of a PPARγ-mediated mechanism in the WSE effects on morphine-mediated nociception and the likely usefulness of WSE in lengthening the analgesic efficacy of opioids in chronic therapy.
Collapse
|
35
|
Greenwald MK. Anti-stress neuropharmacological mechanisms and targets for addiction treatment: A translational framework. Neurobiol Stress 2018; 9:84-104. [PMID: 30238023 PMCID: PMC6138948 DOI: 10.1016/j.ynstr.2018.08.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/30/2018] [Accepted: 08/10/2018] [Indexed: 12/18/2022] Open
Abstract
Stress-related substance use is a major challenge for treating substance use disorders. This selective review focuses on emerging pharmacotherapies with potential for reducing stress-potentiated seeking and consumption of nicotine, alcohol, marijuana, cocaine, and opioids (i.e., key phenotypes for the most commonly abused substances). I evaluate neuropharmacological mechanisms in experimental models of drug-maintenance and relapse, which translate more readily to individuals presenting for treatment (who have initiated and progressed). An affective/motivational systems model (three dimensions: valence, arousal, control) is mapped onto a systems biology of addiction approach for addressing this problem. Based on quality of evidence to date, promising first-tier neurochemical receptor targets include: noradrenergic (α1 and β antagonist, α2 agonist), kappa-opioid antagonist, nociceptin antagonist, orexin-1 antagonist, and endocannabinoid modulation (e.g., cannabidiol, FAAH inhibition); second-tier candidates may include corticotropin releasing factor-1 antagonists, serotonergic agents (e.g., 5-HT reuptake inhibitors, 5-HT3 antagonists), glutamatergic agents (e.g., mGluR2/3 agonist/positive allosteric modulator, mGluR5 antagonist/negative allosteric modulator), GABA-promoters (e.g., pregabalin, tiagabine), vasopressin 1b antagonist, NK-1 antagonist, and PPAR-γ agonist (e.g., pioglitazone). To address affective/motivational mechanisms of stress-related substance use, it may be advisable to combine agents with actions at complementary targets for greater efficacy but systematic studies are lacking except for interactions with the noradrenergic system. I note clinically-relevant factors that could mediate/moderate the efficacy of anti-stress therapeutics and identify research gaps that should be pursued. Finally, progress in developing anti-stress medications will depend on use of reliable CNS biomarkers to validate exposure-response relationships.
Collapse
Affiliation(s)
- Mark K. Greenwald
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, 48201, USA
| |
Collapse
|
36
|
Assessment of pioglitazone and proinflammatory cytokines during buprenorphine taper in patients with opioid use disorder. Psychopharmacology (Berl) 2018; 235:2957-2966. [PMID: 30079432 PMCID: PMC7286070 DOI: 10.1007/s00213-018-4986-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 07/24/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Preliminary evidence suggested that the PPARγ agonist pioglitazone reduces opioid-withdrawal symptoms, possibly by inhibiting increases in proinflammatory cytokines. METHODS A randomized, placebo-controlled clinical trial was conducted utilizing two different study designs (entirely outpatient, and a combination of inpatient and outpatient) to evaluate the safety and efficacy of pioglitazone as an adjunct medication for people with opioid physical dependence undergoing a buprenorphine taper. Participants were stabilized on buprenorphine/naloxone (sublingual, up to 16/4 mg/day), then randomized to receive oral pioglitazone (up to 45 mg/day) or placebo before, during, and after buprenorphine taper. Outcome measures included the Subjective Opiate Withdrawal Scale (SOWS) and Clinical Opiate Withdrawal Scale, use of rescue medications to alleviate opioid withdrawal symptoms, and opioid-positive urine specimens. Cerebrospinal fluid (CSF) and plasma were collected during the taper in a subset of participants for measurement of proinflammatory cytokines. RESULTS The clinical trial was prematurely terminated due to slow enrollment; 40 participants per group were required for adequate statistical power to test study hypotheses. Twenty-four participants enrolled; 17 received at least one dose of study medication (6 pioglitazone, 11 placebo). SOWS scores were higher in the pioglitazone arm than in the placebo arm after adjusting for use of rescue medications; participants in the pioglitazone arm needed more rescue medications than the placebo arm during the post-taper phase. SOWS scores were positively correlated with monocyte chemoattractant protein-1 (MCP-1) in CSF (r = 0.70, p = 0.038) and plasma (r = 0.77, p = 0.015). Participants having higher levels of plasma MCP-1 reported higher SOWS, most notably after the buprenorphine taper ended. CONCLUSIONS Results from this study provide no evidence that pioglitazone reduces opioid withdrawal symptoms during buprenorphine taper. High correlations between MCP-1 and opioid withdrawal symptoms support a role of proinflammatory processes in opioid withdrawal. TRIAL REGISTRATION clinicaltrials.gov identifier: NCT01517165.
Collapse
|
37
|
Jones JD, Bisaga A, Metz VE, Manubay JM, Mogali S, Ciccocioppo R, Madera G, Doernberg M, Comer SD. The PPARγ Agonist Pioglitazone Fails to Alter the Abuse Potential of Heroin, But Does Reduce Heroin Craving and Anxiety. J Psychoactive Drugs 2018; 50:390-401. [PMID: 30204554 DOI: 10.1080/02791072.2018.1508789] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Possibly through its effects on glia, the peroxisome proliferator-activated gamma receptor (PPARγ) agonist pioglitazone (PIO) has been shown to alter the effects of heroin in preclinical models. Until now, these results have not been assessed in humans. Heroin-dependent participants were randomized to either active (45 mg, n = 14) or placebo (0 mg, n = 16) PIO maintenance for the duration of the three-week study. After stabilization on buprenorphine (8 mg), participants began a two-week testing period. On the first to fourth test days, participants could self-administer drug or money by making verbal choices for either option. On the fifth day, active heroin and money were administered and participants could work to receive heroin or money using a progressive ratio choice procedure. Test days 6-10 were identical to test days 1-5 with the exception that, during one of the test weeks, placebo was available on the first four days, and during the other week heroin was available. PIO failed to alter the reinforcing or positive subjective effects of heroin, but it did reduce heroin craving and overall anxiety. Although we were unable to replicate the robust effects found in preclinical models, these data provide an indication of drug effects that deserves further exploration.
Collapse
Affiliation(s)
- Jermaine D Jones
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Adam Bisaga
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Verena E Metz
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Jeanne M Manubay
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Shanthi Mogali
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Roberto Ciccocioppo
- b Department of Experimental Medicine and Public Health, School of Pharmacy, Pharmacology Unit , University of Camerino , Macerata , Italy
| | - Gabriela Madera
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Molly Doernberg
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Sandra D Comer
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| |
Collapse
|
38
|
Jones JD, Comer SD, Metz VE, Manubay JM, Mogali S, Ciccocioppo R, Martinez S, Mumtaz M, Bisaga A. Pioglitazone, a PPARγ agonist, reduces nicotine craving in humans, with marginal effects on abuse potential. Pharmacol Biochem Behav 2017; 163:90-100. [PMID: 29020601 PMCID: PMC5959043 DOI: 10.1016/j.pbb.2017.10.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/20/2017] [Accepted: 10/04/2017] [Indexed: 11/26/2022]
Abstract
Possibly through their actions upon glia, peroxisome proliferator-activated receptor agonists (PPAR) have been shown to alter the abuse potential of addictive drugs in several preclinical models. The current study extends this research into the human laboratory as the first clinical study into the effects of the PPAR gamma agonist, pioglitazone, on the abuse potential of nicotine. Heavy smokers were recruited for this 3-week study. Upon admission, participants were randomized to either active (45mg, n=14) or placebo (0mg, n=13) PIO maintenance conditions for the duration of the study. After 5-7days of stabilization on a 7mg nicotine patch, participants began laboratory testing. On the 1st-4th test days, participants could self-administer cigarettes or receive money by making verbal choices for either option. On the 5th day, participants were administered 10 puffs of their usual brand of cigarette in the morning and later chose between smoking and money by making finger presses on a computer mouse in a progressive ratio self-administration task. Later on the 5th day participants also underwent a smoking cue exposure session. The 8th-11th test days were identical to the 1st-4th test days with the exception that during one of the test weeks de-nicotinized cigarettes were available, and during the other nicotinized cigarettes were available. Nicotinized cigarettes were always administered on the 5th and 12th days. On some measures PIO increased indicators of abuse potential, though this effect was typically not statistically significant. However, PIO did significantly reduce measures of craving.
Collapse
Affiliation(s)
- Jermaine D. Jones
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA,Corresponding author. (J.D. Jones)
| | - Sandra D. Comer
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Verena E. Metz
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Jeanne M. Manubay
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Shanthi Mogali
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, Camerino, Macerata 62032, Italy
| | - Suky Martinez
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA,Translational Research Training Program in Addiction, City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Mudassir Mumtaz
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA,Translational Research Training Program in Addiction, City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Adam Bisaga
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| |
Collapse
|
39
|
Bachtell RK, Jones JD, Heinzerling KG, Beardsley PM, Comer SD. Glial and neuroinflammatory targets for treating substance use disorders. Drug Alcohol Depend 2017; 180:156-170. [PMID: 28892721 PMCID: PMC5790191 DOI: 10.1016/j.drugalcdep.2017.08.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/28/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND The plenary session at the 2016 Behavior, Biology and Chemistry: Translational Research in Addiction Conference focused on glia as potential players in the development, persistence and treatment of substance use disorders. Glia partake in various functions that are important for healthy brain activity. Drugs of abuse alter glial cell activity producing several perturbations in brain function that are thought to contribute to behavioral changes associated with substance use disorders. Consequently, drug-induced changes in glia-driven processes in the brain represent potential targets for pharmacotherapeutics treating substance use disorders. METHODS Four speakers presented preclinical and clinical research illustrating the effects that glial modulators have on abuse-related behavioral effects of psychostimulants and opioids. This review highlights some of these findings and expands its focus to include other research focused on drug-induced glia abnormalities and glia-focused treatment approaches in substance use disorders. RESULTS Preclinical findings show that drugs of abuse induce neuroinflammatory signals and disrupt glutamate homeostasis through their interaction with microglia and astrocytes. Preclinical and clinical studies testing the effects of glial modulators show general effectiveness in reducing behaviors associated with substance use disorders. CONCLUSIONS The contribution of drug-induced glial activity continues to emerge as an intriguing target for substance use disorder treatments. Clinical investigations of glial modulators have yielded promising results on substance use measures and indicate that they are generally safe and well-tolerated. However, results have not been entirely positive and more questions remain for continued exploration in the development and testing of glial-directed treatments for substance use disorders.
Collapse
Affiliation(s)
- Ryan K. Bachtell
- Department of Psychology and Neuroscience, and Center for Neuroscience, UCB 345, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Jermaine D. Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute and College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Keith G. Heinzerling
- Department of Family Medicine and Center for Behavioral and Addiction Medicine, UCLA, Los Angeles, CA, USA
| | - Patrick M. Beardsley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, VA 23298, USA
| | - Sandra D. Comer
- Division on Substance Use Disorders, New York State Psychiatric Institute and College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|