151
|
Ahmed M, Ul Haq I, Faisal M, Waseem D, Taqi MM. Implication of OPRM1 A118G Polymorphism in Opioids Addicts in Pakistan: In vitro and In silico Analysis. J Mol Neurosci 2018; 65:472-479. [PMID: 30033503 PMCID: PMC6132783 DOI: 10.1007/s12031-018-1123-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 07/11/2018] [Indexed: 12/04/2022]
Abstract
Single nucleotide polymorphism in OPRM1 gene is associated with hedonic and reinforcing consequences of opioids. Risk and protective alleles may vary in different populations. One hundred healthy controls and 100 opioids (predominantly heroin) addicts from Pakistani origin were genotyped for A118G (N40D) polymorphism in OPRM1. Structural and functional impact of the polymorphism on encoded protein was predicted by in silico analysis. Results show significant association between homozygous GG genotype and opioid addiction in Pakistani population (p value = 0.016). In silico analysis by SIFT (TI = 0.61), PolyPhen (PISC = 0.227), PANTHER (subPSEC = -1.7171), and SNP effect predicted this SNP benign for encoded protein. Superimposing wild-type and mutated proteins by MODELLER shows no change (RMSD = 0.1) in extracellular ligand binding domain of μ-opioid receptor. However, Haploreg and RegulomeDB predicted OPRM1 gene repression by chromatin condensation and increased binding affinity of RXRA transcription factor that may reduce protein translation and hence the number of available receptors to bind with drugs, which may trigger underlying mechanisms for opioids addiction. Thus, this study outlines causal relationship between opioids addiction and genetic predisposition in Pakistani population.
Collapse
Affiliation(s)
- Madiha Ahmed
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ihsan Ul Haq
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Muhammad Faisal
- Faculty of Health Studies, University of Bradford, Richmond Rd, Bradford, UK.
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK.
| | - Durdana Waseem
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Malik Mumtaz Taqi
- Division of Mental Health and Addiction, NORMENT, University of Oslo, Oslo, Norway
| |
Collapse
|
152
|
Chronic Oxycodone Self-administration Altered Reward-related Genes in the Ventral and Dorsal Striatum of C57BL/6J Mice: An RNA-seq Analysis. Neuroscience 2018; 393:333-349. [PMID: 30059705 DOI: 10.1016/j.neuroscience.2018.07.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/28/2022]
Abstract
Prescription opioid abuse, for example of oxycodone, is a pressing public health issue. This study focuses on how chronic oxycodone self-administration (SA) affects the reward pathways in the mouse brain. In this study, we tested the hypothesis that the expression of reward-related genes in the ventral and dorsal striatum, areas involved in different aspects of opioid addiction models, was altered within 1 h after chronic oxycodone SA, using transcriptome-wide sequencing (RNA-seq). Based on results from earlier human genetic and rodent preclinical studies, we focused on a set of genes that may be associated with the development of addictive diseases and the rewarding effect of drugs of abuse, primarily in the opioid, stress response and classical neurotransmitter systems. We found that 32 transcripts in the ventral striatum, and 7 in the dorsal striatum, were altered significantly in adult mice that had self-administered oxycodone (n = 5) for 14 consecutive days (4 h/day) compared with yoked saline controls (n = 5). The following 5 genes in the ventral striatum showed experiment-wise significant changes: proopiomelanocortin (Pomc) and serotonin 5-HT-2A receptor (Htr2a) were upregulated; serotonin receptor 7 (Htr7), galanin receptor1 (Galr1) and glycine receptor 1 (Glra1) were downregulated. Some genes detected by RNA-seq were confirmed by quantitative polymerase chain reaction (qPCR). Conclusion: A RNA-seq study shows that chronic oxycodone SA alters the expression of several reward-related genes in the dorsal and ventral striatum. These results suggest potential mechanisms underlying neuronal adaptation to chronic oxycodone self-exposure, of relevance to our mechanistic understanding of prescription opioid abuse.
Collapse
|
153
|
Zgheib NK, Aouad MT, Taha SK, Nassar AH, Masri RF, Khoury MY, Makki MH, Siddik-Sayyid SM. μ-opioid receptor genetic polymorphisms and duration of epidural fentanyl analgesia during early labor. Minerva Anestesiol 2018; 84:946-954. [DOI: 10.23736/s0375-9393.18.12697-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
154
|
Zhou Y, Kreek MJ. Involvement of Activated Brain Stress Responsive Systems in Excessive and "Relapse" Alcohol Drinking in Rodent Models: Implications for Therapeutics. J Pharmacol Exp Ther 2018; 366:9-20. [PMID: 29669731 PMCID: PMC5988024 DOI: 10.1124/jpet.117.245621] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 04/16/2018] [Indexed: 02/06/2023] Open
Abstract
Addictive diseases, including addiction to alcohol, pose massive public health costs. Addiction is a chronic relapsing disease caused by both the direct effects induced by drugs and persistent neuroadaptations at the molecular, cellular, and behavioral levels. These drug-type specific neuroadaptations are brought on largely by the reinforcing effects of drugs on the central nervous system and environmental stressors. Results from animal experiments have demonstrated important interactions between alcohol and stress-responsive systems. Addiction to specific drugs such as alcohol, psychostimulants, and opioids shares some common direct or downstream effects on the brain's stress-responsive systems, including arginine vasopressin and its V1b receptors, dynorphin and the κ-opioid receptors, pro-opiomelanocortin/β-endorphin and the μ-opioid receptors, and the endocannabinoids. Further study of these systems through laboratory-based and translational research could lead to the discovery of novel treatment targets and the early optimization of interventions (for example, combination) for the pharmacologic therapy of alcoholism.
Collapse
Affiliation(s)
- Yan Zhou
- Laboratory of Biology of Addictive Diseases, Rockefeller University, New York, New York
| | - Mary Jeanne Kreek
- Laboratory of Biology of Addictive Diseases, Rockefeller University, New York, New York
| |
Collapse
|
155
|
Effects of Single Nucleotide Polymorphisms on Surgical and Postsurgical Opioid Requirements: A Systematic Review and Meta-Analysis. Clin J Pain 2018; 33:1117-1130. [PMID: 28379874 DOI: 10.1097/ajp.0000000000000498] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES There is great heterogeneity in the way individuals respond to medications. Inherited differences, such as single nucleotide polymorphisms (SNP), can influence the efficacy and toxicity of drugs. This meta-analysis aims to collate data from studies investigating the effect of SNPs on postoperative and/or intraoperative opioid requirements. MATERIALS AND METHODS A meta-analysis was conducted following PRISMA guidelines. Eligibility criteria for studies included were reporting amount of postoperative and/or intraoperative opioid used as the primary outcome and genotyping patients for SNPs in one of the following genes; OPRM1, CYP2D6, CYP3A4, CYP3A5, COMT, UGT2B7, or ABCB1. A comprehensive systematic search for articles using keywords "opioid-sensitivity," "polymorphisms," "post-operative opioid," "post-surgical opioid," "post-operative pain," and "post-surgical pain" was performed. RESULTS Fifty-one studies were included. Individuals homozygous for AA at the OPRMI (rs1799971) polymorphisms required less postsurgical opioid compared with those homozygous for GG (Hedges g, -0.270; 95% confidence interval, -0.433 to -0.108; P=0.001). Polymorphisms in CYP2D6, CYP3A4, CYP3A5, COMT, UGT2B7, and ABCB1 did not affect opioid requirements. DISCUSSION Investigation of single changes in 1 gene can only yield limited information regarding genetic effects on opioid requirements. Rapid development of whole genome sequencing enables information on all genetic modifications that may affect analgesic response to be collected. The information collected must include data on the individual's metabolic enzymes, as well as information on drug receptors and enzymes responsible for drug degradation, so that a personal profile can be built up which will predict individual response to drugs, and guide clinicians on the type and dosage of drug to use.
Collapse
|
156
|
Fu Y, Selcuk E, Moore SR, Depue RA. Touch-induced face conditioning is mediated by genetic variation in opioid but not oxytocin receptors. Sci Rep 2018; 8:9004. [PMID: 29899398 PMCID: PMC5998070 DOI: 10.1038/s41598-018-27199-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 05/15/2018] [Indexed: 12/26/2022] Open
Abstract
Soft touch possesses strong prosocial effects that facilitate social bonding and group cohesion in animals. Touch activates opioids (OP) and oxytocin (OXT), two neuromodulators involved in affiliative behaviors and social bonding. We examined whether touch serves as an unconditioned reward in affective conditioning of human faces, a basic process in social bonding, and whether this process is mediated by variation in mu-OP (OPRM1) and OXT (rs53576) receptor genes. Participants viewed affectively-neutral human faces, half of which were paired with a brief soft brushing on the forearm as an unconditioned stimulus (US). Paired and unpaired faces were rated for positive affective and sensory features of touch. Variation in OPRM1 but not rs53576 significantly modulated strength and development of conditioning, indicating that touch-induced mu-OP but not OXT activity provides rewarding properties of a US in conditioning. Implications for touch-induced mu-OP activity in normal and disordered conditioned social bonding are discussed.
Collapse
MESH Headings
- Analgesics, Opioid/metabolism
- Conditioning, Operant/physiology
- Face/physiology
- Female
- Genotype
- Humans
- Male
- Oxytocin/metabolism
- Photic Stimulation
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Receptors, Opioid, mu/physiology
- Receptors, Oxytocin/genetics
- Receptors, Oxytocin/metabolism
- Receptors, Oxytocin/physiology
- Reward
- Social Behavior
- Touch/physiology
- Young Adult
Collapse
Affiliation(s)
- Yu Fu
- Department of Human Development and Institute of Human Neuroscience, Cornell University, Ithaca, New York, 14850, USA
| | - Emre Selcuk
- Department of Human Development and Institute of Human Neuroscience, Cornell University, Ithaca, New York, 14850, USA
- Department of Psychology, Middle East Technical University, Ankara, 06800, Turkey
| | - Sarah R Moore
- Department of Human Development and Institute of Human Neuroscience, Cornell University, Ithaca, New York, 14850, USA
| | - Richard A Depue
- Department of Human Development and Institute of Human Neuroscience, Cornell University, Ithaca, New York, 14850, USA.
| |
Collapse
|
157
|
Zahari Z, Ibrahim MA, Musa N, Tan SC, Mohamad N, Ismail R. Sleep quality and OPRM1 polymorphisms: a cross-sectional study among opioid-naive individuals. BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902018000117217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Zalina Zahari
- Universiti Sultan Zainal Abidin, Malaysia; Universiti Sains Malaysia, Malaysia
| | | | | | | | - Nasir Mohamad
- Universiti Sains Malaysia, Malaysia; Universiti Sultan Zainal Abidin, Malaysia
| | - Rusli Ismail
- Universiti Sains Malaysia, Malaysia; Universiti Sultan Zainal Abidin, Malaysia
| |
Collapse
|
158
|
Kosten TR, Graham DP, Nielsen DA. Neurobiology of Opioid Use Disorder and Comorbid Traumatic Brain Injury. JAMA Psychiatry 2018; 75:642-648. [PMID: 29710079 DOI: 10.1001/jamapsychiatry.2018.0101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
IMPORTANCE Treating patients with opioid use disorder (OUD) and traumatic brain injury illustrates 6 neurobiological principles about the actions of 2 contrasting opioid analgesics, morphine and fentanyl, as well as pharmacotherapies for OUD, methadone, naltrexone, and buprenorphine. OBSERVATIONS This literature review focused on a patient with traumatic brain injury who developed OUD from chronic morphine analgesia. His treatment is described in a neurobiological framework of 6 opioid action principles. CONCLUSIONS AND RELEVANCE The 6 principles are (1) coactivation of neuronal and inflammatory immune receptors (Toll-like receptor 4), (2) 1 receptor activating cyclic adenosine monophosphate and β-arrestin second messenger systems, (3) convergence of opioid and adrenergic receptor types on 1 second messenger, (4) antagonist (eg, naltrexone)-induced receptor trafficking, (5) genetic μ-opioid receptor variants influencing analgesia and tolerance, and (6) cross-tolerance vs receptor antagonism as the basis of OUD pharmacotherapy with methadone or buprenorphine vs naltrexone.
Collapse
Affiliation(s)
- Thomas R Kosten
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - David P Graham
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - David A Nielsen
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| |
Collapse
|
159
|
Rogacki MK, Golfetto O, Tobin SJ, Li T, Biswas S, Jorand R, Zhang H, Radoi V, Ming Y, Svenningsson P, Ganjali D, Wakefield DL, Sideris A, Small AR, Terenius L, Jovanović‐Talisman T, Vukojević V. Dynamic lateral organization of opioid receptors (kappa, mu wt and mu N40D ) in the plasma membrane at the nanoscale level. Traffic 2018; 19:690-709. [PMID: 29808515 PMCID: PMC6120469 DOI: 10.1111/tra.12582] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/06/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023]
Abstract
Opioid receptors are important pharmacological targets for the management of numerous medical conditions (eg, severe pain), but they are also the gateway to the development of deleterious side effects (eg, opiate addiction). Opioid receptor signaling cascades are well characterized. However, quantitative information regarding their lateral dynamics and nanoscale organization in the plasma membrane remains limited. Since these dynamic properties are important determinants of receptor function, it is crucial to define them. Herein, the nanoscale lateral dynamics and spatial organization of kappa opioid receptor (KOP), wild type mu opioid receptor (MOPwt ), and its naturally occurring isoform (MOPN40D ) were quantitatively characterized using fluorescence correlation spectroscopy and photoactivated localization microscopy. Obtained results, supported by ensemble-averaged Monte Carlo simulations, indicate that these opioid receptors dynamically partition into different domains. In particular, significant exclusion from GM1 ganglioside-enriched domains and partial association with cholesterol-enriched domains was observed. Nanodomain size, receptor population density and the fraction of receptors residing outside of nanodomains were receptor-specific. KOP-containing domains were the largest and most densely populated, with the smallest fraction of molecules residing outside of nanodomains. The opposite was true for MOPN40D . Moreover, cholesterol depletion dynamically regulated the partitioning of KOP and MOPwt , whereas this effect was not observed for MOPN40D .
Collapse
Affiliation(s)
- Maciej K. Rogacki
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| | - Ottavia Golfetto
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Steven J. Tobin
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Tianyi Li
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| | - Sunetra Biswas
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Raphael Jorand
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Huiying Zhang
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Vlad Radoi
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| | - Yu Ming
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| | - Per Svenningsson
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| | - Daniel Ganjali
- Department of Mechanical and Aerospace EngineeringThe Henry Samueli School of Engineering, University of CaliforniaIrvineCalifornia
| | - Devin L. Wakefield
- Department of Molecular Medicine, Beckman Research Institute, City of HopeDuarteCalifornia
| | - Athanasios Sideris
- Department of Mechanical and Aerospace EngineeringThe Henry Samueli School of Engineering, University of CaliforniaIrvineCalifornia
| | - Alexander R. Small
- Department of Physics and AstronomyCalifornia State Polytechnic UniversityPomonaCalifornia
| | - Lars Terenius
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
- Department of Molecular and Cellular NeurosciencesThe Scripps Research InstituteLa JollaCalifornia
| | | | - Vladana Vukojević
- Department of Clinical NeuroscienceCenter for Molecular Medicine, Karolinska InstituteStockholmSweden
| |
Collapse
|
160
|
Muriel J, Margarit C, Planelles B, Serralta MJ, Puga C, Inda MDM, Cutillas E, Morales D, Horga JF, Peiró AM. OPRM1 influence on and effectiveness of an individualized treatment plan for prescription opioid use disorder patients. Ann N Y Acad Sci 2018; 1425:82-93. [PMID: 29781244 DOI: 10.1111/nyas.13735] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/16/2018] [Accepted: 03/23/2018] [Indexed: 12/25/2022]
Abstract
Screening for opioid use disorder should be considered in chronic non-cancer pain (CNCP) patients with long-term use of opioids. The aim of our study was to assess the effectiveness of an individualized treatment plan (ITP) for prescription opioid dependence that included screening of pharmacogenetic markers. An observational prospective study was performed using prescription opioid-dependent CNCP outpatients (n = 88). Patients were divided into nonresponders, responders, or high responders according to their response to the ITP. Genotyping of OPRM1 (A118G), OPRD1 (T921C), COMT (G472A), ABCB1 (C3435T), and ARRB2 (C8622T) was performed by real-time PCR. Our ITP achieved a significant reduction of the morphine equivalent daily dose (MEDD) in 64% of responders, including 33% of high responders. Nonopioid medication or buprenorphine use was significantly higher at final versus basal visit. 118-AA OPRM1 patients required significantly lower MEDD at basal and final visits. Our ITP showed effectiveness and security in reducing MEDD in opioid-dependent patients, with good conversion to buprenorphine that was more pronounced in 118-AA OPRM1 patients.
Collapse
Affiliation(s)
- Javier Muriel
- Neuropharmacology on Pain (NED) Research Group, Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante-General Hospital, Alicante, Spain.,Occupational Observatory, Miguel Hernández University of Elche, Elche, Spain
| | - César Margarit
- Neuropharmacology on Pain (NED) Research Group, Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante-General Hospital, Alicante, Spain.,Pain Unit, Department of Health of Alicante-General Hospital, Alicante, Spain
| | - Beatriz Planelles
- Neuropharmacology on Pain (NED) Research Group, Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante-General Hospital, Alicante, Spain.,Pain Unit, Department of Health of Alicante-General Hospital, Alicante, Spain
| | - María J Serralta
- Clinical Psychology and Psychiatry Unit, Department of Health of Alicante-General Hospital, Alicante, Spain
| | - Carmen Puga
- Neuropharmacology on Pain (NED) Research Group, Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante-General Hospital, Alicante, Spain
| | - María-Del-Mar Inda
- Neuropharmacology on Pain (NED) Research Group, Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante-General Hospital, Alicante, Spain
| | - Esperanza Cutillas
- Neuropharmacology on Pain (NED) Research Group, Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante-General Hospital, Alicante, Spain.,Pain Unit, Department of Health of Alicante-General Hospital, Alicante, Spain
| | - Domingo Morales
- Operations Research Centre, Miguel Hernández University of Elche, Elche, Spain
| | - José F Horga
- Clinical Pharmacology Unit, Department of Health of Alicante-General Hospital, Alicante, Spain
| | - Ana M Peiró
- Neuropharmacology on Pain (NED) Research Group, Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante-General Hospital, Alicante, Spain.,Pain Unit, Department of Health of Alicante-General Hospital, Alicante, Spain.,Clinical Pharmacology Unit, Department of Health of Alicante-General Hospital, Alicante, Spain
| |
Collapse
|
161
|
Abstract
Pain is an unpleasant feeling usually resulting from tissue damage that can persist along weeks, months, or even years after the injury, turning into pathological chronic pain, the leading cause of disability. Currently, pharmacology interventions are usually the first-line therapy but there is a highly variable analgesic drug response. Pharmacogenetics (PGx) offers a means to identify genetic biomarkers that can predict individual analgesic response opening doors to precision medicine. PGx analyze the way in which the presence of variations in the DNA sequence (single-nucleotide polymorphisms, SNPs) could be responsible for portions of the population reaching different levels of pain relief (phenotype) due to gene interference in the drug mechanism of action (pharmacodynamics) and/or its concentration at the place of action (pharmacokinetics). SNPs in the cytochrome P450 enzymes genes (CYP2D6) influence metabolism of codeine, tramadol, hydrocodone, oxycodone, and tricyclic antidepressants. Blood concentrations of some NSAIDs depend on CYP2C9 and/or CYP2C8 activity. Additional candidate genes encode for opioid receptors, transporters, and other molecules important for pharmacotherapy in pain management. However, PGx studies are often contradictory, slowing the uptake of this information. This is likely due, in large part, to a lack of robust evidence demonstrating clinical utility and to its polygenic response modulated by other exogenous or epigenetics factors. Novel therapies, including targeting of epigenetic changes and gene therapy-based approaches, broaden future options to improve understanding of pain and the treatment of people who suffer it.
Collapse
Affiliation(s)
- Ana M Peiró
- Clinical Pharmacology Unit, Department of Health of Alicante-General Hospital, Alicante, Spain; Neuropharmacology on Pain (NED), Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante, Spain.
| |
Collapse
|
162
|
Peled I, Yavin E. Predictive Model for the Sequence-Dependent Fluorogenic Response of Forced-Intercalation Peptide Nucleic Acid. ACS OMEGA 2018; 3:3813-3818. [PMID: 30023882 PMCID: PMC6045369 DOI: 10.1021/acsomega.8b00184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/23/2018] [Indexed: 06/08/2023]
Abstract
UNLABELLED The forced-intercalation peptide nucleic acid (FIT-PNA) concept, introduced by Seitz and co-workers, is based on replacing a nucleobase of the PNA sequence with a cyanine dye (such as thiazole orange). The cyanine dye is thus a surrogate base that is forced to intercalate in the duplex (e.g. , PNA DNA). This allows single-mismatch sensitivity as the introduction of a mismatch in the vicinity of the dye increases freedom of motion and leads to a significant depletion of its fluorescence because of the free rotation of the monomethine bond separating the two π-systems of the cyanine dye. Herein, we designed and synthesized six FIT-PNA probes, featuring bisquinoline (BisQ), a red-emitting cyanine dye recently developed in our laboratory for FIT-PNAs. By following PNA-DNA duplex fluorescence, we found new sequence-based factors governing the fluorescence response to the mismatched FIT-PNA:DNA duplex. Fluorogenic properties are correlated with the π-stacking energy of three distinctive base pair steps (BPSs) in the PNA:DNA duplex. The first two are the two BPSs opposite BisQ, whereas the third is the BPS of the mismatch position, which presumably becomes unstacked due to the mismatch. We suggest a predictive model for FIT-PNA single-mismatch detection mechanism, a model that can be used in future research to improve FIT-PNA design.
Collapse
Affiliation(s)
| | - Eylon Yavin
- E-mail: . Phone: +972-2-6758692. Fax: +972-2-6757574 (E.Y.)
| |
Collapse
|
163
|
Gao X, Wang Y, Lang M, Yuan L, Reece AS, Wang W. Contribution of Genetic Polymorphisms and Haplotypes in DRD2, BDNF, and Opioid Receptors to Heroin Dependence and Endophenotypes Among the Han Chinese. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 21:404-412. [PMID: 28692418 DOI: 10.1089/omi.2017.0057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Heroin and drug dependence are major contributors to global health burden worldwide, but their underlying mechanisms remain elusive and may vary from population to population. Reward- and memory-related candidate genes dopamine D2 receptor (DRD2) and brain-derived neurotrophic factor (BDNF), as well as the opioid receptor genes (OPRM1, OPRD1, and OPRK1), have been implicated in drug dependence, but relatively little is known on their contributions to heroin dependence in populations worldwide. Hence, we evaluated the contributions of the above five candidate genes in heroin dependence and several important related endophenotypes (the onset age of heroin use and subjective response to first heroin use), at single single-nucleotide polymorphism as well as haplotype levels, in a Han Chinese population sample. We genotyped 546 unrelated and heroin-dependent subjects for the candidate genes noted, and 228 sex- and age-matched unrelated controls. The G allele of rs4654327 (OPRD1), DRD2 haplotype block CCGCCGTT (rs6277-rs1076560-rs2283265-rs2734833-rs2075652-rs1079596-rs4436578-rs11214607), and OPRD1 haplotypes TACG (rs6669447-rs2236857-rs508448-rs4654327), CG (rs508448-rs4654327), and TG (rs6669447-rs4654327) were significantly associated with heroin dependence phenotype. Homozygotes AA at rs6265 (BDNF), TT at rs16917234 (BDNF), and CC at rs508448 (OPRD1) also appeared as risk factors for the endophenotype earlier age of onset for heroin use. Two OPRM1 haplotypes, AG (rs1799971-rs1381376) and AT (rs1799971-rs3778151), were observed as potential protective factors. These emerging findings contribute to the literature on genetic biomarkers of drug dependence and related endophenotypes, and call for replication in independent population.
Collapse
Affiliation(s)
- Xuan Gao
- 1 Savaid Medical School, University of Chinese Academy of Sciences , Beijing, P.R. China
| | - Youxin Wang
- 2 School of Public Health, Capital Medical University , Beijing, P.R. China
| | - Minglin Lang
- 3 College of Life Science, University of Chinese Academy of Sciences , Beijing, P.R. China
| | - Li Yuan
- 1 Savaid Medical School, University of Chinese Academy of Sciences , Beijing, P.R. China
| | - Albert Stuart Reece
- 4 Divison of Psychiatry, University of Western Australia , Crawley, Australia
| | - Wei Wang
- 2 School of Public Health, Capital Medical University , Beijing, P.R. China .,5 School of Medical and Health Sciences, Edith Cowan University , Perth, Australia
| |
Collapse
|
164
|
Henderson-Redmond AN, Lowe TE, Tian XB, Morgan DJ. Increased ethanol drinking in "humanized" mice expressing the mu opioid receptor A118G polymorphism are mediated through sex-specific mechanisms. Brain Res Bull 2018; 138:12-19. [PMID: 28780411 PMCID: PMC5796878 DOI: 10.1016/j.brainresbull.2017.07.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 11/22/2022]
Abstract
The A118G single nucleotide polymorphism (SNP) of the mu-opioid receptor gene (Oprm1) has been implicated in mediating the rewarding effects of alcohol. Clinical and preclinical studies suggest that the G allele may confer a genetic vulnerability to alcohol dependence, though it remains unknown whether these effects are sex-specific. We used male and female mice homozygous for the "humanized" 118AA or 118GG alleles to determine whether the A118G SNP potentiates ethanol consumption in a sex-specific manner in both the two-bottle choice and drinking-in-the-dark (DID) paradigms. Mice were also assessed for differences in naltrexone sensitivity, ethanol reward assessed via conditioned place preference (CPP), and sensitivity to the sedative/ataxic effects of ethanol using the rota-rod and loss of righting reflex (LORR) assays. We found that male and female 118GG mice drank significantly more ethanol than 118AA littermates using a continuous access, two-bottle choice paradigm. In the limited-access DID drinking model, (i) female (but not male) 118GG mice consumed more ethanol than 118AA mice and (ii) naltrexone pretreatment was equally efficacious at attenuating ethanol intake in both 118AA and 118GG female mice while having no effect in males. Male and female 118GG and female 118AA mice developed a robust conditioned place preference (CPP) for ethanol. Female 118GG mice displayed less sensitivity to the sedative/ataxic effects of ethanol compared to female 118AA mice on both the rota-rod and the LORR assays while male mice did not differ in their responses on either assay. Our findings suggest that increased ethanol consumption in male 118GG mice may be due to increased ethanol reward, while increased drinking in female 118GG mice might be due to decreased sensitivity to the sedative/ataxic effects of ethanol. Collectively, these data might be used to help identify sex-specific pharmacotherapies to combat alcohol use disorders.
Collapse
MESH Headings
- Alcohol Drinking/genetics
- Alcohol Drinking/physiopathology
- Alleles
- Analgesics, Non-Narcotic/pharmacology
- Analysis of Variance
- Animals
- Choice Behavior/drug effects
- Conditioning, Operant/drug effects
- Dose-Response Relationship, Drug
- Ethanol/administration & dosage
- Ethanol/blood
- Female
- Genotype
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutagenesis, Site-Directed
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Polymorphism, Single Nucleotide/genetics
- Quinine/pharmacology
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Reflex/drug effects
- Reflex/genetics
- Reward
- Self Administration
- Self Stimulation
- Sex Characteristics
Collapse
Affiliation(s)
- Angela N Henderson-Redmond
- Department of Anesthesiology & Perioperative Medicine, Penn State University College of Medicine, Hershey, PA 17033, United States.
| | - Tammy E Lowe
- Department of Anesthesiology & Perioperative Medicine, Penn State University College of Medicine, Hershey, PA 17033, United States; Benedict College, Columbia, SC 29204, United States
| | - Xi B Tian
- Department of Anesthesiology & Perioperative Medicine, Penn State University College of Medicine, Hershey, PA 17033, United States
| | - Daniel J Morgan
- Department of Anesthesiology & Perioperative Medicine, Penn State University College of Medicine, Hershey, PA 17033, United States; Department of Pharmacology, Penn State University College of Medicine, Hershey, PA 17033, United States; Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey PA 17033, United States.
| |
Collapse
|
165
|
Li DJ, Chung KS, Wu HC, Hsu CY, Yen CF. Factors affecting the dose of methadone among patients receiving methadone maintenance therapy in Taiwan. Am J Addict 2018; 27:225-230. [DOI: 10.1111/ajad.12712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 11/27/2022] Open
Affiliation(s)
- Dian-Jeng Li
- Department of Addiction Science; Kaohsiung Municipal Kai-Syuan Psychiatric; Hospital; Kaohsiung Taiwan
- Graduate Institute of Medicine; College of Medicine; Kaohsiung Medical University; Kaohsiung Taiwan
| | - Kuan-Shang Chung
- Department of Addiction Science; Kaohsiung Municipal Kai-Syuan Psychiatric; Hospital; Kaohsiung Taiwan
| | - Hung-Chi Wu
- Department of Addiction Science; Kaohsiung Municipal Kai-Syuan Psychiatric; Hospital; Kaohsiung Taiwan
| | - Chih-Yao Hsu
- Department of Addiction Science; Kaohsiung Municipal Kai-Syuan Psychiatric; Hospital; Kaohsiung Taiwan
| | - Cheng-Fang Yen
- Graduate Institute of Medicine; College of Medicine; Kaohsiung Medical University; Kaohsiung Taiwan
- Department of Psychiatry; Kaohsiung Medical University Hospital; Kaohsiung Taiwan
| |
Collapse
|
166
|
Fine-Tuning Limited Proteolysis: A Major Role for Regulated Site-Specific O-Glycosylation. Trends Biochem Sci 2018; 43:269-284. [PMID: 29506880 DOI: 10.1016/j.tibs.2018.02.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/26/2018] [Accepted: 02/02/2018] [Indexed: 11/23/2022]
Abstract
Limited proteolytic processing is an essential and ubiquitous post-translational modification (PTM) affecting secreted proteins; failure to regulate the process is often associated with disease. Glycosylation is also a ubiquitous protein PTM and site-specific O-glycosylation in close proximity to sites of proteolysis can regulate and direct the activity of proprotein convertases, a disintegrin and metalloproteinases (ADAMs), and metalloproteinases affecting the activation or inactivation of many classes of proteins, including G-protein-coupled receptors (GPCRs). Here, we summarize the emerging data that suggest O-glycosylation to be a key regulator of limited proteolysis, and highlight the potential for crosstalk between multiple PTMs.
Collapse
|
167
|
Xie W, Zhuang W, Chen L, Xie W, Jiang C, Liu N. 4218T/C polymorphism associations with post-cesarean patient-controlled epidural fentanyl consumption and pain perception. Acta Anaesthesiol Scand 2018; 62:376-383. [PMID: 29148033 DOI: 10.1111/aas.13040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/03/2017] [Accepted: 10/29/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND The utilization of intrathecal opioids is an efficacious component of post-cesarean section pain management. Given that growing evidence indicates that calcitonin gene-related peptide (CGRP) plays a key role in the development of peripheral sensitization and is associated with enhanced pain, we hypothesized that CGRP 4218T/C polymorphism is associated with the variability in fentanyl consumption for post-cesarean analgesia. METHODS We recruited 548 patients who presented for elective cesarean delivery, and used polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method to analyze CGRP 4218T/C polymorphism. We examined the association of CGRP 4218T/C polymorphism and post-operative fentanyl consumption for analgesia as well as adverse reactions to fentanyl in those patients who received cesarean section surgeries. RESULTS We found that the CGRP 4218T/C polymorphism has a significant effect on pain perception, analgesic requirement, and nausea and vomiting for the first 24 h after cesarean delivery in patients who received PCEA fentanyl. Individuals with the C/C genotype had more pain, required more PCEA fentanyl, and experienced a lower incidence of nausea and vomiting. CONCLUSION These results indicated that patients with C/C genotype may have reduced sensitivity to fentanyl analgesia and/or increased pain perception, and were more willing to use PCEA fentanyl to manage their pain.
Collapse
Affiliation(s)
- W. Xie
- Department of Anesthesiology; Quanzhou First Hospital; Quanzhou China
| | - W. Zhuang
- Department of Anesthesiology; Huian Hospital; Quanzhou China
| | - L. Chen
- Department of Anesthesiology; Huian Hospital; Quanzhou China
| | - W. Xie
- Department of Anesthesiology; Quanzhou First Hospital; Quanzhou China
| | - C. Jiang
- Department of Anesthesiology; Quanzhou First Hospital; Quanzhou China
| | - N. Liu
- Department of Anesthesiology; Quanzhou First Hospital; Quanzhou China
| |
Collapse
|
168
|
Soleimani Asl S, Roointan A, Bergen H, Amiri S, Mardani P, Ashtari N, Shabani R, Mehdizadeh M. Opioid Receptors Gene Polymorphism and Heroin Dependence in Iran. Basic Clin Neurosci 2018; 9:101-106. [PMID: 29967669 PMCID: PMC6026094 DOI: 10.29252/nirp.bcn.9.2.101] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Introduction: Genes often have multiple polymorphisms that interact with each other and the environment in different individuals. Variability in the opioid receptors can influence opiate withdrawal and dependence. In humans, A118G Single Nucleotide Polymorphisms (SNP) on μ-Opioid Receptor (MOR), 36 G>T in κ-Opioid Receptor (KOR), and T921C in the δ-Opioid Receptor (DOR) have been found to associate with substance dependence. Methods: To investigate the association between opioid receptors gene polymorphism and heroin addiction, 100 control subjects with no history of opioid use, and 100 heroin addicts (50% males and 50% females) in Tehran (capital of Iran), were evaluated. A118G, 36 G>T, and T921C SNPs on the MOR, KOR, DOR genes, respectively, were genotyped by sequencing. Results: We found no differences in either allele or genotype frequency for MOR, KOR and DOR genes SNPs between controls and subjects addicted to heroin. Conclusion: The relationships among polymorphisms may be important in determining the risk profile for complex diseases such as addiction, but opioid addiction is a multifactorial syndrome which is partially hereditary and partially affected by the environment.
Collapse
Affiliation(s)
- Sara Soleimani Asl
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir Roointan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hugo Bergen
- Department of Human Anatomy and Cell Sciences, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shayan Amiri
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parastoo Mardani
- Department of Biology, Faculty of Science, Tehran Branch, Payame Noor University, Tehran, Iran
| | - Niloufar Ashtari
- Department of Human Anatomy and Cell Sciences, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ronak Shabani
- Cellular and Molecular Research Center, Department of Anatomy, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mehdizadeh
- Cellular and Molecular Research Center, Department of Anatomy, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
169
|
Politi C, Ciccacci C, Novelli G, Borgiani P. Genetics and Treatment Response in Parkinson's Disease: An Update on Pharmacogenetic Studies. Neuromolecular Med 2018; 20:1-17. [PMID: 29305687 DOI: 10.1007/s12017-017-8473-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 12/29/2017] [Indexed: 01/11/2023]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder characterized by a progressive loss of dopamine neurons of the central nervous system. The disease determines a significant disability due to a combination of motor symptoms such as bradykinesia, rigidity and rest tremor and non-motor symptoms such as sleep disorders, hallucinations, psychosis and compulsive behaviors. The current therapies consist in combination of drugs acting to control only the symptoms of the illness by the replacement of the dopamine lost. Although patients generally receive benefits from this symptomatic pharmacological management, they also show great variability in drug response in terms of both efficacy and adverse effects. Pharmacogenetic studies highlighted that genetic factors play a relevant influence in this drug response variability. In this review, we tried to give an overview of the recent progresses in the pharmacogenetics of PD, reporting the major genetic factors identified as involved in the response to drugs and highlighting the potential use of some of these genomic variants in the clinical practice. Many genes have been investigated and several associations have been reported especially with adverse drug reactions. However, only polymorphisms in few genes, including DRD2, COMT and SLC6A3, have been confirmed as associated in different populations and in large cohorts. The identification of genomic biomarkers involved in drug response variability represents an important step in PD treatment, opening the prospective of more personalized therapies in order to identify, for each person, the better therapy in terms of efficacy and toxicity and to improve the PD patients' quality of life.
Collapse
Affiliation(s)
- Cristina Politi
- Department of Biomedicine and Prevention, Genetics Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Cinzia Ciccacci
- Department of Biomedicine and Prevention, Genetics Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy.
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Genetics Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Paola Borgiani
- Department of Biomedicine and Prevention, Genetics Section, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| |
Collapse
|
170
|
Yuferov V, Butelman ER, Kreek MJ. Gender-specific association of functional prodynorphin 68 bp repeats with cannabis exposure in an African American cohort. Neuropsychiatr Dis Treat 2018; 14:1025-1034. [PMID: 29713172 PMCID: PMC5909790 DOI: 10.2147/ndt.s159954] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cannabis use disorders (CUDs) cause substantial neuropsychiatric morbidity and comorbidity. There is evidence for gender-based differences in CUDs, for instance, a greater prevalence in males than in females. The main active component of cannabis is delta 9-tetrahydrocannabinol (delta 9-THC), a partial agonist of the cannabinoid type 1 receptor. Preclinical studies show that genetic or pharmacological manipulation of the kappa opioid receptor/dynorphin system modulates the effects of delta 9-THC. METHODS In this case-control study of adult African Americans (n=476; 206 females, 270 males), we examined the association of the functional prodynorphin 68 bp (PDYN 68 bp) promoter repeats with categorical diagnoses of cannabis dependence (Diagnostic and Statistical Manual of Mental Disorders-IV criteria), as well as with a rapid dimensional measure of maximum lifetime cannabis exposure (the Kreek-McHugh-Schluger-Kellogg cannabis scale). RESULTS The PDYN 68 bp genotype (examined as short-short [SS], short-long [SL], or long-long [LL], based on the number of repeats) was not significantly associated with categorical cannabis-dependence diagnoses, either in males or in females. However, in males, the PDYN 68 bp SS+SL genotype was associated with both greater odds of any use of cannabis (p<0.05) and earlier age of first cannabis use, compared to the LL genotype (ie, 15 versus 16.5 years of age; p<0.045). Males in the SS+SL group also had greater odds of high lifetime exposure to cannabis, compared to the LL group (p<0.045). Of interest, none of the aforementioned genetic associations were significant in females. CONCLUSION This study provides the first data on how the PDYN 68 bp genotype is associated with gender-specific patterns of exposure to cannabis. Overall, this study shows that PDYN 68 bp polymorphisms affect behaviors involved in early stages of nonmedical cannabis use and potentially lead to increasing self-exposure. These data may eventually lead to improvements in personalized medicine for the prevention and treatment of highly prevalent CUDs and neuropsychiatric comorbidities.
Collapse
Affiliation(s)
- Vadim Yuferov
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Eduardo R Butelman
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| |
Collapse
|
171
|
Butelman ER, Maremmani AGI, Bacciardi S, Chen CY, Correa da Rosa J, Kreek MJ. Non-medical Cannabis Self-Exposure as a Dimensional Predictor of Opioid Dependence Diagnosis: A Propensity Score Matched Analysis. Front Psychiatry 2018; 9:283. [PMID: 29997535 PMCID: PMC6030387 DOI: 10.3389/fpsyt.2018.00283] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022] Open
Abstract
Background: The impact of increasing non-medical cannabis use on vulnerability to develop opioid use disorders has received considerable attention, with contrasting findings. A dimensional analysis of self-exposure to cannabis and other drugs, in individuals with and without opioid dependence (OD) diagnoses, may clarify this issue. Objective: To examine the age of onset of maximal self-exposure to cannabis, alcohol, cocaine, and heroin, in volunteers diagnosed with OD, using a rapidly administered instrument (the KMSK scales). To then determine whether maximal self-exposure to cannabis, alcohol, and cocaine is a dimensional predictor of odds of OD diagnoses. Methods: This outpatient observational study examined maximal self-exposure to these drugs, in volunteers diagnosed with DSM-IV OD or other drug diagnoses, and normal volunteers. In order to focus more directly on opioid dependence diagnosis as the outcome, volunteers who had cocaine dependence diagnoses were excluded. Male and female adults of diverse ethnicity were consecutively ascertained from the community, and from local drug treatment programs, in 2002-2013 (n = 574, of whom n = 94 had OD diagnoses). The age of onset of maximal self-exposure of these drugs was examined. After propensity score matching for age at ascertainment, gender, and ethnicity, a multiple logistic regression examined how increasing self-exposure to non-medical cannabis, alcohol and cocaine affected odds of OD diagnoses. Results: Volunteers with OD diagnoses had the onset of heaviest use of cannabis in the approximate transition between adolescence and adulthood (mean age = 18.9 years), and onset of heaviest use of alcohol soon thereafter (mean age = 20.1 years). Onset of heaviest use of heroin and cocaine was detected later in the lifespan (mean ages = 24.7 and 25.3 years, respectively). After propensity score matching for demographic variables, we found that the maximal self-exposure to cannabis and cocaine, but not to alcohol, was greater in volunteers with OD diagnoses, than in those without this diagnosis. Also, a multiple logistic regression detected that increasing self-exposure to cannabis and cocaine, but not alcohol, was a positive predictor of OD diagnosis. Conclusions/Importance: Increasing self-exposure to non-medical cannabis, as measured with a rapid dimensional instrument, was a predictor of greater odds of opioid dependence diagnosis, in propensity score-matched samples.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, Rockefeller University, New York, NY, United States
| | - Angelo G I Maremmani
- "VP Dole" Dual Diagnosis Unit, Azienda Ospedaliera-Universitaria Pisana, Pisa, Italy
| | - Silvia Bacciardi
- "VP Dole" Dual Diagnosis Unit, Azienda Ospedaliera-Universitaria Pisana, Pisa, Italy
| | - Carina Y Chen
- Laboratory on the Biology of Addictive Diseases, Rockefeller University, New York, NY, United States
| | - Joel Correa da Rosa
- Center for Clinical and Translational Science, The Rockefeller University Hospital, Rockefeller University, New York, NY, United States
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, Rockefeller University, New York, NY, United States
| |
Collapse
|
172
|
Butelman ER, Kreek MJ. Discriminative Stimulus Properties of Opioid Ligands: Progress and Future Directions. Curr Top Behav Neurosci 2018; 39:175-192. [PMID: 27225498 DOI: 10.1007/7854_2016_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Opioid receptors (MOP-r, KOP-r, DOP-r, as well as NOP-r) and their endogenous neuropeptide agonist systems are involved in diverse neurobiological and behavioral functions, in health and disease. These functions include pain and analgesia, addictions, and psychiatric diseases (e.g., depression-, anxiety-like, and stress-related disorders). Drug discrimination assays have been used to characterize the behavioral pharmacology of ligands with affinity at MOP-r, KOP-r, or DOP-r (and to a lesser extent NOP-r). Therefore, drug discrimination studies with opioid ligands have an important continuing role in translational investigations of diseases that are affected by these neurobiological targets and their pharmacotherapy.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, Box 171, New York, NY, 10065, USA.
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, Box 171, New York, NY, 10065, USA
| |
Collapse
|
173
|
Nerenz RD, Tsongalis GJ. Pharmacogenetics of Opioid Use and Implications for Pain Management. J Appl Lab Med 2018; 2:622-632. [PMID: 33636885 DOI: 10.1373/jalm.2017.023150] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/05/2017] [Indexed: 11/06/2022]
Abstract
BACKGROUND Opioid analgesics are frequently prescribed to manage acute and chronic pain, but individual differences in opioid response make effective pain control in all patients an elusive goal. Furthermore, the risk of addiction following opioid consumption varies among individual patients. Although many psychosocial factors contribute to an individual's opioid response and risk for addiction, a strong genetic component has also been demonstrated. CONTENT Opioids undergo substantial enzymatic modification that can generate metabolites with either increased or decreased opioid activity relative to the parent compound. To elicit their analgesic effect, parent compounds and active metabolites must be transported into the central nervous system where they bind to opioid receptors and inhibit neurotransmission. Inherited genetic variants that alter the function of proteins involved in these processes have been associated with differences in opioid response and risk for addiction. Detection of these variants can help guide opioid selection, inform dosing decisions, or encourage use of a nonopioid analgesic. SUMMARY Whereas some genetic variants are clearly associated with differences in opioid response and have been included in consensus clinical practice guidelines, the impact of other variants on opioid response remains unclear. Studies performed to date have generated promising results, but inconsistent findings, reimbursement challenges, and the lack of robust decision support tools have hampered widespread adoption of pharmacogenetic testing to guide pain management treatment decisions. Future work involving the simultaneous evaluation of large numbers of variants and demonstration of a clear clinical benefit provided by pharmacogenetic testing will be required to overcome these obstacles.
Collapse
Affiliation(s)
- Robert D Nerenz
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH.,The Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Gregory J Tsongalis
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH.,The Geisel School of Medicine at Dartmouth, Hanover, NH
| |
Collapse
|
174
|
Collins D, Randesi M, da Rosa JC, Zhang Y, Kreek MJ. Oprm1 A112G, a single nucleotide polymorphism, alters expression of stress-responsive genes in multiple brain regions in male and female mice. Psychopharmacology (Berl) 2018; 235:2703-2711. [PMID: 30027498 PMCID: PMC6132675 DOI: 10.1007/s00213-018-4965-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/02/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND OPRM1 A118G, a functional human mu-opioid receptor (MOR) polymorphism, is associated with drug dependence and altered stress responsivity in humans as well as altered MOR signaling. MOR signaling can regulate many cellular processes, including gene expression, and many of the long-term, stable effects of drugs and stress may stem from changes in gene expression in diverse brain regions. A mouse model bearing an equivalent polymorphism (Oprm1 A112G) was previously generated and studied. Mice homozygous for the G112 allele show differences in opioid- and stress-related phenotypes. APPROACH The current study examines the expression of 24 genes related to drug and stress responsivity in the caudoputamen, nucleus accumbens, hypothalamus, hippocampus, and amygdala of drug-naïve, stress-minimized, male and female mice homozygous for either the G112 variant allele or the wild-type A112 allele. RESULTS We detected nominal genotype-dependent changes in gene expression of multiple genes. We also detected nominal sex-dependent as well as sex-by-genotype interaction effects on gene expression. Of these, four genotype-dependent differences survived correction for multiple testing: Avp and Gal in the hypothalamus and Oprl1 and Cnr1 in the hippocampus. CONCLUSIONS Changes in the regulation of these genes by mu-opioid receptors encoded by the G112 allele may be involved in some of the behavioral and molecular consequences of this polymorphism observed in mice.
Collapse
Affiliation(s)
- Devon Collins
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA.
| | - Matthew Randesi
- 0000 0001 2166 1519grid.134907.8The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Joel Correa da Rosa
- 0000 0001 2166 1519grid.134907.8Laboratory of Investigative Dermatology, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Yong Zhang
- 0000 0001 2166 1519grid.134907.8The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Mary Jeanne Kreek
- 0000 0001 2166 1519grid.134907.8The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| |
Collapse
|
175
|
Putative Independent Evolutionary Reversals from Genotypic to Temperature-Dependent Sex Determination are Associated with Accelerated Evolution of Sex-Determining Genes in Turtles. J Mol Evol 2017; 86:11-26. [DOI: 10.1007/s00239-017-9820-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 11/18/2017] [Indexed: 12/14/2022]
|
176
|
Predictors of Naltrexone Response in a Randomized Trial: Reward-Related Brain Activation, OPRM1 Genotype, and Smoking Status. Neuropsychopharmacology 2017; 42:2640-2653. [PMID: 28409564 PMCID: PMC5686497 DOI: 10.1038/npp.2017.74] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 03/01/2017] [Accepted: 04/04/2017] [Indexed: 01/14/2023]
Abstract
Naltrexone reduces drinking among individuals with alcohol use disorders (AUDs), but it is not effective for everyone. Variability in its effects on reward-related brain activation, genetic variation, and/or cigarette smoking may account for this mixed response profile. This randomized clinical trial tested the effects of naltrexone on drinking and alcohol cue-elicited brain activation, evaluated whether OPRM1 A118G genotype or smoking moderated these effects, and explored whether the effects of medication on cue-elicited activation predicted subsequent drinking. One hundred and fifty-two treatment-seeking individuals with alcohol dependence, half preselected to carry at least one A118G G (Asp) allele, were randomized to naltrexone (50 mg) or placebo for 16 weeks and administered an fMRI alcohol cue reactivity task at baseline and after 2 weeks of treatment. Naltrexone, relative to placebo, significantly reduced alcohol cue-elicited activation of the right ventral striatum (VS) between baseline and week 2 and reduced heavy drinking over 16 weeks. OPRM1 genotype did not significantly moderate these effects, but G-allele carriers who received naltrexone had an accelerated return to heavy drinking after medication was stopped. Smoking moderated the effects of medication on drinking, such that naltrexone was superior to placebo only among smokers. The degree of reduction in right VS activation between scans interacted with medication in predicting subsequent drinking, such that individuals with greater reduction in activation who received naltrexone, but not placebo, experienced the least heavy drinking during the following 14 weeks. These data replicate previous findings that naltrexone reduces heavy drinking and reward-related brain activation among treatment-seeking individuals with AUDs, and indicate that smoking and the magnitude of reduction in cue-elicited brain activation may predict treatment response.
Collapse
|
177
|
Abstract
AbstractAnimal models of early postnatal mother–infant interactions have highlighted the importance of tactile contact for biobehavioral outcomes via the modification of DNA methylation (DNAm). The role of normative variation in contact in early human development has yet to be explored. In an effort to translate the animal work on tactile contact to humans, we applied a naturalistic daily diary strategy to assess the link between maternal contact with infants and epigenetic signatures in children 4–5 years later, with respect to multiple levels of child-level factors, including genetic variation and infant distress. We first investigated DNAm at four candidate genes: the glucocorticoid receptor gene, nuclear receptor subfamily 3, group C, member 1 (NR3C1), μ-opioid receptor M1 (OPRM1) and oxytocin receptor (OXTR; related to the neurobiology of social bonds), and brain-derived neurotrophic factor (BDNF; involved in postnatal plasticity). Although no candidate gene DNAm sites significantly associated with early postnatal contact, when we next examined DNAm across the genome, differentially methylated regions were identified between high and low contact groups. Using a different application of epigenomic information, we also quantified epigenetic age, and report that for infants who received low contact from caregivers, greater infant distress was associated with younger epigenetic age. These results suggested that early postnatal contact has lasting associations with child biology.
Collapse
|
178
|
|
179
|
Ragia G, Veresies I, Veresie L, Veresies K, Manolopoulos VG. Association study of DRD2 A2/A1, DRD3 Ser9Gly, DβH -1021C>T, OPRM1 A118G and GRIK1 rs2832407C>A polymorphisms with alcohol dependence. Drug Metab Pers Ther 2017; 31:143-50. [PMID: 27447243 DOI: 10.1515/dmpt-2016-0015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 06/24/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND The reinforcing effects of alcohol are mediated through complex interactions between multiple neurochemical systems. Genes of dopaminergic (DRD2, DRD3 and DβH), opioid (OPRM1) and glutaminergic (GRIK1) systems mediate the dependent behavior via different mechanisms; however, they all target the serotonergic and dopaminergic pathways in the ventral tegmental area. METHODS In the present study, DRD2 A2/A1, DRD3 Ser9Gly, DβH -1021C>T, OPRM1 A118G and GRIK1 rs2832407C>A polymorphisms and their interactions were analyzed in 72 alcohol-dependent patients and 74 controls of Greek-Cypriot origin, using the PCR-RFLP method. RESULTS No differences were found in the genotype or allele distribution of DRD2 A2/A1, DRD3 Ser9Gly, DβH -1021C>T, OPRM1 A118G and GRIK1 rs2832407C>A between alcohol-dependent patients and controls. Additionally, we did not find any gene×gene interactions in association with alcohol dependence in the studied population. CONCLUSIONS Alcohol dependence is a complex interaction of genetic and environmental factors. In the present study, we have shown that DRD2 A2/A1, DRD3 Ser9Gly, DβH -1021C>T, OPRM1 A118G and GRIK1 rs2832407C>A are not associated with this dependent behavior alone or in interaction.
Collapse
|
180
|
Peiró AM, Planelles B, Juhasz G, Bagdy G, Libert F, Eschalier A, Busserolles J, Sperlagh B, Llerena A. Pharmacogenomics in pain treatment. Drug Metab Pers Ther 2017; 31:131-42. [PMID: 27662648 DOI: 10.1515/dmpt-2016-0005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/08/2016] [Indexed: 11/15/2022]
Abstract
The experience of chronic pain is one of the commonest reasons for seeking medical attention, being a major issue in clinical practice. While pain is a universal experience, only a small proportion of people who felt pain develop pain syndromes. In addition, painkillers are associated with wide inter-individual variability in the analgesic response. This may be partly explained by the presence of single nucleotide polymorphisms in genes encoding molecular entities involved in pharmacodynamics and pharmacokinetics. However, uptake of this information has been slow due in large part to the lack of robust evidences demonstrating clinical utility. Furthermore, novel therapies, including targeting of epigenetic changes and gene therapy-based approaches are further broadening future options for the treatment of chronic pain. The aim of this article is to review the evidences behind pharmacogenetics (PGx) to individualize therapy (boosting the efficacy and minimizing potential toxicity) and genes implicated in pain medicine, in two parts: (i) genetic variability with pain sensitivity and analgesic response; and (ii) pharmacological concepts applied on PGx.
Collapse
|
181
|
Abstract
Pain sensitivity is an inherited factor that varies strongly between individuals. We investigated whether genetic polymorphisms in the candidate genes COMT, OPRM1, OPRD1, TAOK3, TRPA1, TRPV1, and SCN9A are contributing to experimental pain variability between children. Our study included 136 children and adolescents (8-18 years). Cold and heat pain thresholds were determined with a Thermal Sensory Analyzer. Women and young children were significantly more sensitive to pain (P < 0.05). After correction for age, gender, reaction time, and correction for multiple testing, OPRM1 118A>G G-allele carriers (AG and GG) rated the hot stimulus as significantly less painful than did OPRM1 118A>G AA genotyped individuals (2[1-5] vs 7 [3-9], respectively; P = 0.00005). Additionally, OPRM1 118G allele carriers reached more frequently the minimum temperature limit (44% vs 17%, respectively; P = 0.003) and maximum temperature limit (52% vs 24%, respectively; P = 0.0052), indicative for lower pain sensitivity. The combined genotype, based on expected pain sensitivity, OPRM1 118AA/COMT 472 GA or AA genotyped children, was associated with lower pain thresholds (ie, higher pain sensitivity) than were the OPRM1 118GA or GG/COMT 472GG genotyped children. This is the first study reporting on genetic variants and experimental thermal pain in children and adolescents. OPRM1 rs1799971 and the combined OPRM1/COMT genotype could serve as biomarkers for pain sensitivity.
Collapse
|
182
|
Driscoll CA, Lindell SG, Schwandt ML, Suomi SJ, Higley JD, Heilig M, Barr CS. OPRM1 genotype interacts with serotonin system dysfunction to predict alcohol-heightened aggression in primates. Addict Biol 2017; 22:1655-1664. [PMID: 27484010 DOI: 10.1111/adb.12428] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 06/10/2016] [Accepted: 06/21/2016] [Indexed: 11/28/2022]
Abstract
Although the notion that alcohol promotes violence is widespread, not all individuals are aggressive while intoxicated. Genetic variation could be a contributing factor to individual differences in alcohol-heightened aggression. The present study examines the effects of OPRM1C77G genotype on responses to threat in rhesus macaques under normal conditions and following alcohol administration. Prior studies have shown that a low CSF level of 5-HIAA is a trait marker for individuals prone to escalated aggression. We wanted to examine whether the predictive value for this marker on aggression was moderated by OPRM1 genotype. Animals were administered alcohol (BAC 100-200 mg%), were provoked by a human intruder, and aggressive responses were recorded. Factor analysis was performed to generate aggressive response factors, which were then used as dependent variables for ANOVA, with OPRM1 genotype and CSF 5-HIAA as independent variables. Factor analysis generated three factors ('Threatening', 'Distance Decreasing' and 'High Intensity'). We found that High Intensity aggression was increased among carriers of the OPRM1 G allele, especially among individuals with low CSF levels of 5-HIAA. Aggression in the non-intoxicated state was predicted by 5-HIAA, but not by genotype. This study demonstrates that OPRM1 genotype predicts alcohol-heightened aggression in rhesus macaques with low CSF levels of 5-HIAA. Because OPRM1 variation predicts similar effects on alcohol response and behavior in humans and macaques, this study could suggest a role for OPRM1 genotype in alcohol-heightened aggression in humans. If so, it may be that compounds that block this receptor could reduce alcohol-associated violence in selected patient populations.
Collapse
Affiliation(s)
- Carlos A Driscoll
- Section of Comparative Behavioral Genomics; LNG/NIAAA/NIH; Rockville MD United States
| | - Stephen G. Lindell
- Section of Comparative Behavioral Genomics; LNG/NIAAA/NIH; Rockville MD United States
| | - Melanie L Schwandt
- Laboratory of Clinical and Translational Studies; NIAAA/NIH; Bethesda MD United States
| | - Stephen J Suomi
- Laboratory of Comparative Ethology; NIH/NICHD; Poolesville MD United States
| | - J Dee Higley
- Department of Psychology; Brigham Young University; Provo UT United States
| | - Markus Heilig
- Laboratory of Clinical and Translational Studies; NIAAA/NIH; Bethesda MD United States
| | - Christina S Barr
- Section of Comparative Behavioral Genomics; LNG/NIAAA/NIH; Rockville MD United States
| |
Collapse
|
183
|
Korucuoglu O, Gladwin TE, Baas F, Mocking RJ, Ruhé HG, Groot PF, Wiers RW. Neural response to alcohol taste cues in youth: effects of the OPRM1 gene. Addict Biol 2017; 22:1562-1575. [PMID: 27594419 DOI: 10.1111/adb.12440] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 07/19/2016] [Accepted: 07/22/2016] [Indexed: 11/28/2022]
Abstract
Genetic variations in the mu-opioid receptor (OPRM1) gene have been related to high sensitivity to rewarding effects of alcohol. The current study focuses on the neural circuitry underlying this phenomenon using an alcohol versus water taste-cue reactivity paradigm in a young sample at relatively early stages of alcohol use, thus limiting the confound of variations in duration of alcohol use. Drinkers (17-21 years old) were selected on genotype carrying the AA-(n = 20) or the AG-(n = 16) variant of the A118G single nucleotide polymorphism (SNP) of the OPRM1 gene (rs1799971), and underwent functional magnetic resonance imaging (fMRI). Magnitude of the neural activity and frontostriatal functional connectivity in response to alcohol versus water were investigated. The AG-group demonstrated reduced activation in prefrontal and parietal regions, including the inferior and middle frontal gyrus, superior and inferior parietal lobule, compared with the AA-group. No activation differences were observed in the mesolimbic pathway. Connectivity from the ventral-striatum to frontal regions for alcohol > water trials was higher in the AG than the AA group. For the dorsal-striatum seed region, the AG group showed increased connectivity to non-PFC regions. These results indicate that adolescents carrying the G-allele may be more vulnerable for the alcohol to hijack the reward system in the absence of frontal control to regulate craving. This implies that findings of hyperactivation in the mesolimbic structures of G-allele carriers in earlier studies might result from both genetic susceptibility and heavy drinking.
Collapse
Affiliation(s)
- Ozlem Korucuoglu
- Addiction, Development and Psychopathology (ADAPT)-lab, Department of Psychology; University of Amsterdam; The Netherlands
- Department of Psychological Sciences; University of Missouri; Columbia MO USA
| | - Thomas E. Gladwin
- Addiction, Development and Psychopathology (ADAPT)-lab, Department of Psychology; University of Amsterdam; The Netherlands
- Research Centre - Military Mental Health; Ministry of Defense; The Netherlands
| | - Frank Baas
- Department of Genome Analysis; Academic Medical Centre; The Netherlands
| | - Roel J.T. Mocking
- Program for Mood Disorders, Department of Psychiatry, Academic Medical Center; University of Amsterdam; The Netherlands
| | - Henricus G. Ruhé
- Program for Mood Disorders, Department of Psychiatry, Academic Medical Center; University of Amsterdam; The Netherlands
- University of Groningen, University Medical Center Groningen, Mood and Anxiety Disorders; Department of Psychiatry; The Netherlands
| | - Paul F.C. Groot
- Department of Radiology; Academic Medical Center; The Netherlands
| | - Reinout W. Wiers
- Addiction, Development and Psychopathology (ADAPT)-lab, Department of Psychology; University of Amsterdam; The Netherlands
| |
Collapse
|
184
|
Park JH, Kim SG, Kim JH, Lee JS, Jung WY, Kim HK. Spicy Food Preference and Risk for Alcohol Dependence in Korean. Psychiatry Investig 2017; 14:825-829. [PMID: 29209387 PMCID: PMC5714725 DOI: 10.4306/pi.2017.14.6.825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/25/2017] [Accepted: 06/09/2017] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Previous studies have reported that both preference for spicy food and drinking behavior are associated with the activity of the opioid system in the central nervous system. The relationship between the preference for spicy food and the risk of alcohol dependence by comparing spicy food preference in alcohol-dependent patients vs. healthy controls was investigated. Also the association between the preference for spicy food and OPRM1 A118G was studied. METHODS A total of 150 Korean male patients with alcohol dependence and 100 normal male control subjects were included in this study. Preference for spicy food was measured using the Food Preference Scale (FPS). DNA analysis was conducted to detect the A118G polymorphism. RESULTS The mean FPS score was significantly higher in the alcohol-dependent patients (61.2±24.2) than in the normal control subjects (53.0±22.0). FPS scores differed significantly between alcohol-dependent patients and normal control subjects who had the G allele in OPRM1 A118G, but not between the two groups with the AA genotype. CONCLUSION A strong preference for spicy food can be assumed to be a risk factor for alcohol dependence, particularly in those carrying the G allele in OPRM1 A118G.
Collapse
Affiliation(s)
- Ji-Hun Park
- Department of Psychiatry, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Sung-Gon Kim
- Department of Psychiatry, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Ji-Hoon Kim
- Department of Psychiatry, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Jin-Seong Lee
- Department of Psychiatry, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Woo-Young Jung
- Department of Psychiatry, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Hyeon-Kyeong Kim
- Medical Research Institute, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
185
|
Yeung EW, Craggs JG, Gizer IR. Comorbidity of Alcohol Use Disorder and Chronic Pain: Genetic Influences on Brain Reward and Stress Systems. Alcohol Clin Exp Res 2017; 41:1831-1848. [PMID: 29048744 DOI: 10.1111/acer.13491] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/25/2017] [Indexed: 01/10/2023]
Abstract
Alcohol use disorder (AUD) is highly comorbid with chronic pain (CP). Evidence has suggested that neuroadaptive processes characterized by reward deficit and stress surfeit are involved in the development of AUD and pain chronification. Neurological data suggest that shared genetic architecture associated with the reward and stress systems may contribute to the comorbidity of AUD and CP. This monograph first delineates the prevailing theories of the development of AUD and pain chronification focusing on the reward and stress systems. It then provides a brief summary of relevant neurological findings followed by an evaluation of evidence documented by molecular genetic studies. Candidate gene association studies have provided some initial support for the genetic overlap between AUD and CP; however, these results must be interpreted with caution until studies with sufficient statistical power are conducted and replications obtained. Genomewide association studies have suggested a number of genes (e.g., TBX19, HTR7, and ADRA1A) that are either directly or indirectly related to the reward and stress systems in the AUD and CP literature. Evidence reviewed in this monograph suggests that shared genetic liability underlying the comorbidity between AUD and CP, if present, is likely to be complex. As the advancement in molecular genetic methods continues, future studies may show broader central nervous system involvement in AUD-CP comorbidity.
Collapse
Affiliation(s)
- Ellen W Yeung
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri.,Institute for Interdisciplinary Salivary Bioscience Research, University of California at Irvine, Irvine, California
| | - Jason G Craggs
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri.,School of Health Professions, University of Missouri, Columbia, Missouri
| | - Ian R Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
186
|
Levran O, Peles E, Randesi M, da Rosa JC, Adelson M, Kreek MJ. The μ-opioid receptor nonsynonymous variant 118A>G is associated with prolonged abstinence from heroin without agonist treatment. Pharmacogenomics 2017; 18:1387-1391. [PMID: 28976288 DOI: 10.2217/pgs-2017-0092] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
AIM This study assesses whether opioid-related gene variants contribute to reduced vulnerability to relapse to heroin in persons who are not treated with μ-opioid receptor agonist. METHODS Genotypes of 71 SNPs, in nine genes, were analyzed for association with long-term abstinence in former heroin-dependents of European/Middle Eastern ancestry, either without agonist treatment (n = 129) or in methadone maintenance treatment (n = 922). RESULTS The functional OPRM1 nonsynonymous SNP rs1799971 (118A>G) showed significant association with long-term abstinence (Ppermutation = 0.03, dominant model, OR: 2.2; 95% CI: 1.5-3.3). CONCLUSION Since the stress axis is regulated in part by β-endorphin, this functional OPRM1 SNP may blunt the endogenous stress response and contribute to reduced vulnerability for relapse.
Collapse
Affiliation(s)
- Orna Levran
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, NY, 10065, USA
| | - Einat Peles
- Dr Miriam & Sheldon G Adelson Clinic for Drug Abuse Treatment & Research, Tel Aviv Elias Sourasky Medical Center, 1 Henrietta Szold St, Tel-Aviv, 64924, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, 69978, Israel
| | - Matthew Randesi
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, NY, 10065, USA
| | - Joel Correa da Rosa
- Center for Clinical & Translational Science, The Rockefeller University, NY, 10065, USA
| | - Miriam Adelson
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, NY, 10065, USA.,Dr Miriam & Sheldon G Adelson Clinic for Drug Abuse Treatment & Research, Tel Aviv Elias Sourasky Medical Center, 1 Henrietta Szold St, Tel-Aviv, 64924, Israel.,Dr Miriam & Sheldon G Adelson Clinic for Drug Abuse Treatment & Research, Las Vegas, NV, 89169, USA
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, NY, 10065, USA
| |
Collapse
|
187
|
Kringel D, Ultsch A, Zimmermann M, Jansen JP, Ilias W, Freynhagen R, Griessinger N, Kopf A, Stein C, Doehring A, Resch E, Lötsch J. Emergent biomarker derived from next-generation sequencing to identify pain patients requiring uncommonly high opioid doses. THE PHARMACOGENOMICS JOURNAL 2017; 17:419-426. [PMID: 27139154 PMCID: PMC5637232 DOI: 10.1038/tpj.2016.28] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 11/05/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022]
Abstract
Next-generation sequencing (NGS) provides unrestricted access to the genome, but it produces 'big data' exceeding in amount and complexity the classical analytical approaches. We introduce a bioinformatics-based classifying biomarker that uses emergent properties in genetics to separate pain patients requiring extremely high opioid doses from controls. Following precisely calculated selection of the 34 most informative markers in the OPRM1, OPRK1, OPRD1 and SIGMAR1 genes, pattern of genotypes belonging to either patient group could be derived using a k-nearest neighbor (kNN) classifier that provided a diagnostic accuracy of 80.6±4%. This outperformed alternative classifiers such as reportedly functional opioid receptor gene variants or complex biomarkers obtained via multiple regression or decision tree analysis. The accumulation of several genetic variants with only minor functional influences may result in a qualitative consequence affecting complex phenotypes, pointing at emergent properties in genetics.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/therapeutic use
- Biomarkers, Pharmacological/analysis
- Chronic Pain/drug therapy
- Chronic Pain/genetics
- Dose-Response Relationship, Drug
- Genotype
- High-Throughput Nucleotide Sequencing
- Humans
- Pharmacogenomic Testing
- Pharmacogenomic Variants
- Receptors, Opioid/genetics
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, mu/genetics
- Receptors, sigma/genetics
- Sigma-1 Receptor
Collapse
Affiliation(s)
- D Kringel
- Institute of Clinical Pharmacology, Goethe-University, Frankfurt am Main, Germany
| | - A Ultsch
- DataBionics Research Group, University of Marburg, Marburg, Germany
| | - M Zimmermann
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - W Ilias
- Department of Anaesthesiology and Intensive Care Medicine, Vienna, Austria
| | - R Freynhagen
- Zentrum für Anästhesiologie, Intensivmedizin, Schmerztherapie & Palliativmedizin, Benedictus Krankenhaus Tutzing, Tutzing, Germany
- Klinik für Anästhesiologie, Technische Universität München, München, Germany
| | - N Griessinger
- Department of Anesthesiology, University Hospital Erlangen, Erlangen, Germany
| | - A Kopf
- Department of Anesthesiology and Critical Care Medicine, Freie Universität Berlin–Charité, Berlin, Germany
| | - C Stein
- Department of Anesthesiology and Critical Care Medicine, Freie Universität Berlin–Charité, Berlin, Germany
| | - A Doehring
- Institute of Clinical Pharmacology, Goethe-University, Frankfurt am Main, Germany
| | - E Resch
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology TMP, Frankfurt am Main, Germany
| | - J Lötsch
- Institute of Clinical Pharmacology, Goethe-University, Frankfurt am Main, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology TMP, Frankfurt am Main, Germany
| |
Collapse
|
188
|
Freet CS, Alexander DN, Imperio CG, Ruiz-Velasco V, Grigson PS. Heroin-induced suppression of saccharin intake in OPRM1 A118G mice. Brain Res Bull 2017; 138:73-79. [PMID: 28939474 DOI: 10.1016/j.brainresbull.2017.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/12/2017] [Accepted: 09/15/2017] [Indexed: 12/19/2022]
Abstract
The single nucleotide polymorphism of the μ-opioid receptor, OPRM1 A118G, has been associated with greater drug and alcohol use, increased sensitivity to pain, and reduced sensitivity to the antinociceptive effects of opiates. In the present studies, we employed a 'humanized' mouse model containing the wild-type (118AA) or variant (118GG) allele to examine behavior in a model of heroin-induced devaluation of an otherwise palatable saccharin cue when repeated saccharin-heroin pairings occurred every 24h (Experiment 1) or every 48h (Experiment 2). The results showed that, while both the 118AA and 118GG mice demonstrated robust avoidance of the heroin-paired saccharin cue following daily taste-drug pairings, only the 118AA mice suppressed intake of the heroin-paired saccharin cue when 48h elapsed between each taste-drug pairing. Humanized 118GG mice, then, defend their intake of the sweet cue despite saccharin-heroin pairings and this effect is illuminated by the use of spaced, rather than massed, trials. Given that this pattern of strain difference is not evident with saccharin-cocaine pairings (Freet et al., 2015), reduced avoidance of the heroin-paired saccharin cue by the 118GG mice may be due to an interaction between the opiate and the subjects' drive for the sweet or, alternatively, to differential downstream sensitivity to the aversive kappa mediated properties of the drug. These alternative hypotheses are addressed.
Collapse
Affiliation(s)
- Christopher S Freet
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, United States.
| | - Danielle N Alexander
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Caesar G Imperio
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Victor Ruiz-Velasco
- Department of Anesthesiology and Perioperative Medicine, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Patricia S Grigson
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| |
Collapse
|
189
|
Kongara K. Pharmacogenetics of opioid analgesics in dogs. J Vet Pharmacol Ther 2017; 41:195-204. [DOI: 10.1111/jvp.12452] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 07/28/2017] [Indexed: 02/03/2023]
Affiliation(s)
- K. Kongara
- Animal Welfare Science and Bioethics Centre; Institute of Veterinary, Animal and Biomedical Sciences, Massey University; Palmerston North New Zealand
| |
Collapse
|
190
|
Al-Mustafa MM, Al Oweidi AS, Al-Zaben KR, Qudaisat IY, Abu-Halaweh SA, Al-Ghanem SM, Massad IM, Samarah WK, Al-Shaer RA, Ismail SI. Remifentanil consumption in septoplasty surgery under general anesthesia. Association with humane mu-opioid receptor gene variants. Saudi Med J 2017; 38:170-175. [PMID: 28133690 PMCID: PMC5329629 DOI: 10.15537/smj.2017.2.16348] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Objectives: To evaluate the influence of the ORM1 variants in codon 118 on the intra-operative remifentanil consumption under general anesthesia. Methods: A prospective gene association study, performed at the Jordan University Jordan, Amman, Jordan from September 2013 to August 2014. It includes patients who underwent septoplasty surgery under general anesthesia. All patients received standard intravenous anesthesia. Anesthesia maintained with fixed dose of Sevoflurane and variable dose of Remifentanil to keep the systolic blood pressure between 90-100 mm Hg. The Remifentanil dose was calculated and correlated with ORM1 genotype variance. Results: Genotype and clinical data were available for 123 cases. The A118A genotype was seen in 96 patients (78%), the A118G genotype was seen in 25 patients (20.3%), and only 2 patients had genotype G118G (1.6%). The G118G variant was removed from the statistical analysis due to small sample size. There was a significant effect of ORM1 genotype variant and the amount of remifentanil consumed. The A118A genotype received 0.173 ± 0.063 µg kg-1 min-1 and the A118G genotype received 0.316 ± 0.100 µg kg-1 min-1 (p<0.0001). Conclusion: The ORM1 gene has a role in intra-operative remifentanil consumption in patients who underwent septoplasty surgery under general anesthesia. The A118G gene required higher dose of remifentanil compared with the A118A genotype.
Collapse
Affiliation(s)
- Mahmoud M Al-Mustafa
- Department of Anesthesia and Intensive Care, Jordan University Hospital, The University of Jordan, Amman, Jordan. E-mail.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Hirasawa-Fujita M, Bly MJ, Ellingrod VL, Dalack GW, Domino EF. Genetic Variation of the Mu Opioid Receptor (OPRM1) and Dopamine D2 Receptor (DRD2) is Related to Smoking Differences in Patients with Schizophrenia but not Bipolar Disorder. CLINICAL SCHIZOPHRENIA & RELATED PSYCHOSES 2017; 11:39-48. [PMID: 28548579 PMCID: PMC4366347 DOI: 10.3371/1935-1232-11.1.39] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
It is not known why mentally ill persons smoke excessively. Inasmuch as endogenous opioid and dopaminergic systems are involved in smoking reinforcement, it is important to study mu opioid receptor (OPRM1) A118G (rs1799971), dopamine D2 receptor (DRD2) Taq1A (rs1800497) genotypes, and sex differences among patients with schizophrenia or bipolar disorder. Smokers and nonsmokers with schizophrenia (n=177) and bipolar disorder (n=113) were recruited and genotyped. They were classified into three groups: current smoker, former smoker, and never smoker by tobacco smoking status self-report. The number of cigarettes smoked per day was used as the major tobacco smoking parameter. In patients with schizophrenia, tobacco smoking prevalence was greater in males than in females as expected, but women had greater daily cigarette consumption (p<0.01). Subjects with schizophrenia who had the OPRM1 *G genotype smoked more cigarettes per day than the AA allele carriers with schizophrenia (p<0.05). DRD2 Taq1A genotype differences had no effect on the number of cigarettes smoked per day. However, female smokers with schizophrenia who were GG homozygous of the DRD2 receptor smoked more than the *A male smokers with schizophrenia (p<0.05). In bipolar patients, there were no OPRM1 and DRD2 Taq1A genotype differences in smoking status. There also were no sex differences for smoking behavior among the bipolar patients. The results of this study indicate that single nucleotide polymorphism (SNP) of the less functional mu opioid receptor increases tobacco smoking in patients with schizophrenia. Alteration of DRD2 receptor function also increased smoking behavior in females with schizophrenia.
Collapse
|
192
|
T394A Mutation at the μ Opioid Receptor Blocks Opioid Tolerance and Increases Vulnerability to Heroin Self-Administration in Mice. J Neurosci 2017; 36:10392-10403. [PMID: 27707973 DOI: 10.1523/jneurosci.0603-16.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023] Open
Abstract
The etiology and pathophysiology underlying opioid tolerance and dependence are still unknown. Because mu opioid receptor (MOR) plays an essential role in opioid action, many vulnerability-related studies have focused on single nucleotide polymorphisms of MOR, particularly on A118G. In this study, we found that a single-point mutation at the MOR T394 phosphorylation site could be another important susceptive factor in the development of opioid tolerance and dependence in mice. T394A mutation, in which a threonine at 394 was replaced by an alanine, did not alter agonist binding to MOR and opioid analgesia, but resulted in loss of etorphine-induced MOR internalization in spinal dorsal horn neurons and opioid analgesic tolerance induced by either morphine or etorphine. In addition, this mutation also caused an increase in intravenous heroin self-administration and in nucleus accumbens dopamine response to heroin. These findings suggest that T394 phosphorylation following MOR activation causes MOR internalization and desensitization, which subsequently contributes to the development of tolerance in both opioid analgesia and opioid reward. Accordingly, T394A mutation blocks opioid tolerance and leads to an increase in brain dopamine response to opioids and in opioid-taking behavior. Thus, the T394 may serve as a new drug target for modulating opioid tolerance and the development of opioid abuse and addiction. SIGNIFICANCE STATEMENT The mechanisms underlying opioid tolerance and susceptibility to opioid addiction remain unclear. The present studies demonstrate that a single-point mutation at the T394 phosphorylation site in the C-terminal of mu opioid receptor (MOR) results in loss of opioid tolerance and enhanced vulnerability to heroin self-administration. These findings suggest that modulation of the MOR-T394 phosphorylation or dephosphorylation status may have therapeutic potential in management of pain, opioid tolerance, and opioid abuse and addiction. Accordingly, MOR-T394 mutation or polymorphisms could be a risk factor in developing opioid abuse and addiction and therefore be used as a new biomarker in prediction and prevention of opioid abuse and addiction.
Collapse
|
193
|
|
194
|
Sweeney CG, Rando JM, Panas HN, Miller GM, Platt DM, Vallender EJ. Convergent Balancing Selection on the Mu-Opioid Receptor in Primates. Mol Biol Evol 2017; 34:1629-1643. [PMID: 28333316 PMCID: PMC6279279 DOI: 10.1093/molbev/msx105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The mu opioid receptor is involved in many natural processes including stress response, pleasure, and pain. Mutations in the gene also have been associated with opiate and alcohol addictions as well as with responsivity to medication targeting these disorders. Two common and mutually exclusive polymorphisms have been identified in humans, A118G (N40D), found commonly in non-African populations, and C17T (V6A), found almost exclusively in African populations. Although A118G has been studied extensively for associations and in functional assays, C17T is much less well understood. In addition to a parallel polymorphism previously identified in rhesus macaques (Macaca mulatta), C77G (P26R), resequencing in additional non-human primate species identifies further common variation: C140T (P47L) in cynomolgus macaques (Macaca fascicularis), G55C (D19H) in vervet monkeys (Chlorocebus aethiops sabeus), A111T (L37F) in marmosets (Callithrix jacchus), and C55T (P19S) in squirrel monkeys (Saimiri boliviensis peruviensis). Functional effects on downstream signaling are observed for each of these variants following treatment with the endogenous agonist β-endorphin and the exogenous agonists morphine, DAMGO ([d-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin), and fentanyl. In addition to demonstrating the importance of functional equivalency in reference to population variation for minority health, this also shows how common evolutionary pressures have produced similar phenotypes across species, suggesting a shared response to environmental needs and perhaps elucidating the mechanism by which these organism-environment interactions are mediated physiologically and molecularly. These studies set the stage for future investigations of shared functional polymorphisms across species as a new genetic tool for translational research.
Collapse
Affiliation(s)
- Carolyn G. Sweeney
- Division of Neuroscience, New England Primate Research Center, Harvard Medical School, Southborough, MA
| | - Juliette M. Rando
- Division of Neuroscience, New England Primate Research Center, Harvard Medical School, Southborough, MA
| | - Helen N. Panas
- Division of Neuroscience, New England Primate Research Center, Harvard Medical School, Southborough, MA
| | - Gregory M. Miller
- Division of Neuroscience, New England Primate Research Center, Harvard Medical School, Southborough, MA
| | - Donna M. Platt
- Division of Neuroscience, New England Primate Research Center, Harvard Medical School, Southborough, MA
| | - Eric J. Vallender
- Division of Neuroscience, New England Primate Research Center, Harvard Medical School, Southborough, MA
| |
Collapse
|
195
|
Butelman ER, Bacciardi S, Maremmani AGI, Darst-Campbell M, Correa da Rosa J, Kreek MJ. Can a rapid measure of self-exposure to drugs of abuse provide dimensional information on depression comorbidity? Am J Addict 2017; 26:632-639. [PMID: 28654734 DOI: 10.1111/ajad.12578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/28/2017] [Accepted: 06/04/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Addictions to heroin or to cocaine are associated with substantial psychiatric comorbidity, including depression. Poly-drug self-exposure (eg, to heroin, cocaine, cannabis, or alcohol) is also common, and may further affect depression comorbidity. METHODS This case-control study examined the relationship of exposure to the above drugs and depression comorbidity. Participants were recruited from methadone maintenance clinics, and from the community. Adult male and female participants (n = 1,201) were ascertained consecutively by experienced licensed clinicians. The instruments used were the SCID-I, and Kreek-McHugh-Schluger-Kellogg (KMSK) scales, which provide a rapid dimensional measure of maximal lifetime self-exposure to each of the above drugs. This measure ranges from no exposure to high unit dose, high frequency, and long duration of exposure. RESULTS A multiple logistic regression with stepwise variable selection revealed that increasing exposure to heroin or to cocaine was associated greater odds of depression, with all cases and controls combined. In cases with an opioid dependence diagnosis, increasing cocaine exposure was associated with a further increase in odds of depression. However, in cases with a cocaine dependence diagnosis, increasing exposure to either cannabis or alcohol, as well as heroin, was associated with a further increase in odds of depression. DISCUSSION AND CONCLUSIONS This dimensional analysis of exposure to specific drugs provides insights on depression comorbidity with addictive diseases, and the impact of poly-drug exposure. SCIENTIFIC SIGNIFICANCE A rapid analysis of exposure to drugs of abuse reveals how specific patterns of drug and poly-drug exposure are associated with increasing odds of depression. This approach detected quantitatively how different patterns of poly-drug exposure can result in increased odds of depression comorbidity, in cases diagnosed with opioid versus cocaine dependence. (Am J Addict 2017;26:632-639).
Collapse
Affiliation(s)
- Eduardo Roque Butelman
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Silvia Bacciardi
- "VP Dole" Dual Diagnosis Unit, Santa Chiara University Hospital of Pisa, Pisa, Italy
| | | | - Maya Darst-Campbell
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Joel Correa da Rosa
- Center for Clinical and Translational Science, The Rockefeller University Hospital, New York, New York
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| |
Collapse
|
196
|
Kolliopoulou A, Stratopoulos A, Siamoglou S, Sgourou A, Ali BR, Papachatzopoulou A, Katsila T, Patrinos GP. Key Pharmacogenomic Considerations for Sickle Cell Disease Patients. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2017; 21:314-322. [PMID: 28486096 DOI: 10.1089/omi.2017.0058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Sickle cell disease (SCD), although a monogenic disease, exhibits a complex clinical phenotype that hampers optimum patient stratification and disease management, especially on hydroxyurea treatment. Moreover, theranostics, the combination of diagnostics to individualize and optimize therapeutic interventions, has not been firmly on the forefront of SCD research and clinical management to date. We suggest that if tailor-made theranostics in SCD is envisaged, pharmacogenomics is anticipated to be the way forward. Herein, we present the current key pharmacogenomic opportunities and challenges in SCD, considering population variation, ethics, and socioeconomic aspects. We focus on pharmacogenomics and pain management, genethics, and cost-effectiveness in SCD. We searched for and synthesized data from PubMed and Google Scholar, and the references from relevant articles, using the keywords "pharmacogenomics," "sickle cell disease," "hydroxyurea," "ethics," "pain management," "morphine metabolism," "opioids," "pharmacogenomics and chronic pain," "cost-effectiveness," and "economic evaluation." Only articles published in English were included. So far, when pharmacogenomics in SCD has been considered, interindividual variability in hydroxyurea response/toxicity has been of primary interest. We underscore the need to extend pharmacogenomic considerations on other therapeutic interventions currently present using a holistic patient-centric approach, and taking disease complications into account as well. Furthermore, we raise awareness toward socioeconomic, ethical, and population differences in the way sickle cell pharmacogenomics might unfold in the future. If pharmacogenomics in SCD is to be used in the clinic in an evidence-based manner, cost-effectiveness and population-specific empirical ethics data are urgently needed.
Collapse
Affiliation(s)
- Alexandra Kolliopoulou
- 1 Department of Pharmacy, School of Health Sciences, University of Patras , Patras, Greece
| | - Apostolos Stratopoulos
- 1 Department of Pharmacy, School of Health Sciences, University of Patras , Patras, Greece
| | - Stavroula Siamoglou
- 1 Department of Pharmacy, School of Health Sciences, University of Patras , Patras, Greece
| | | | - Bassam R Ali
- 3 Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University , Al-Ain, United Arab Emirates
| | | | - Theodora Katsila
- 1 Department of Pharmacy, School of Health Sciences, University of Patras , Patras, Greece
| | - George P Patrinos
- 1 Department of Pharmacy, School of Health Sciences, University of Patras , Patras, Greece
- 3 Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University , Al-Ain, United Arab Emirates
| |
Collapse
|
197
|
Cannabinoid Receptor Type 1 and mu-Opioid Receptor Polymorphisms Are Associated With Cyclic Vomiting Syndrome. Am J Gastroenterol 2017; 112:933-939. [PMID: 28349993 DOI: 10.1038/ajg.2017.73] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 02/01/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Cyclic vomiting syndrome (CVS) is a disorder defined by recurrent, unexplained episodes of severe nausea and vomiting. Our aim was to investigate whether CVS and pathophysiological mechanisms underlying this condition are associated with selected variations in genes encoding the components of the endogenous cannabinoid and opioid systems. METHODS This case-control study included 65 patients with CVS-16 male and 49 female, and 1,092 healthy controls-525 male and 567 female from the 1000 Genomes Project. CVS subjects filled out study-specific questionnaires. Single-nucleotide polymorphisms (SNPs) in genes encoding cannabinoid receptors (CNR1 and CNR2), fatty acid amide hydrolase (FAAH) and mu-opioid receptor (OPRM1) were analyzed using the TaqMan SNP genotyping assay. Correlations between SNP's and clinical characteristics of CVS were ascertained. RESULTS Our study disclosed an increased risk of CVS among individuals with AG and GG genotypes of CNR1 rs806380 (P<0.01), whereas the CC genotype of CNR1 rs806368 and AG and GG genotypes of OPRM1 rs1799971 were associated with a decreased risk of CVS (P<0.05). In addition, AG and GG genotypes of OPRM1 rs1799971 were correlated with migraine episodes, AG and GG of OPRM1 rs1799971, and CT and CC of CNR1 rs806368 with a family history of migraines (second degree relatives), and CT and CC of CNR1 rs2023239 with a positive response to therapy. CONCLUSIONS Our results show for the first time that the variations in CNR1 and OPRM1 genes are associated with CVS and that different genotypes may contribute to the risk of CVS.
Collapse
|
198
|
Otto JM, Gizer IR, Deak JD, Fleming KA, Bartholow BD. A cis-eQTL in OPRM1 is Associated with Subjective Response to Alcohol and Alcohol Use. Alcohol Clin Exp Res 2017; 41:929-938. [PMID: 28273335 PMCID: PMC5404990 DOI: 10.1111/acer.13369] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/25/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND A functional polymorphism within the μ-opioid receptor (OPRM1) gene, rs1799971 (A118G), previously has been associated with measures of alcohol use and sensitivity to its effects, but findings have been inconclusive. A recent study suggested that a second nearby variant within OPRM1, rs3778150, is robustly associated with heroin dependence and fully explained a smaller observed association with rs1799971. Given evidence that the rs3778150-C allele is associated with decreased OPRM1 expression levels in the human brain, the current study sought to test the hypothesis that rs3778150 represents a causal variant within OPRM1 that increases risk for a variety of alcohol use phenotypes. METHODS Participants with genotype and phenotype data from a larger experimental study (N = 152) were assessed on measures of subjective response to alcohol and alcohol use. Measures included (i) the Self-Rating of the Effects of Alcohol and the Alcohol Sensitivity Questionnaire, (ii) the Biphasic Alcohol Effects Scale (BAES) and ratings of subjective intoxication, and (iii) average number of drinks per week in the past month. RESULTS Compared to rs3778150-T homozygous individuals, carriers of the rs3778150-C allele exhibited significantly lower retrospective self-report levels of alcohol sensitivity. Carriers of the rs3778150-C allele also exhibited lower levels of BAES alcohol-related stimulation during an alcohol challenge and reported higher levels of drinking in the last 30 days. With the exception of lower levels of BAES alcohol-related sedation, the rs1799971 variant did not show consistent significant association with any of the alcohol phenotypes in the presence of rs3778150. CONCLUSIONS Results suggest that rs3778150 may be causally related to alcohol use phenotypes, and could potentially account for previously observed associations of rs1799971 with substance use phenotypes. Future studies may investigate potential causal relations among genetic variants in OPRM1, subjective response to alcohol, and drinking phenotypes to further delineate the effects of rs3778150.
Collapse
Affiliation(s)
- Jacqueline M Otto
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri
| | - Ian R Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri
| | - Joseph D Deak
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri
| | - Kimberly A Fleming
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri
| | - Bruce D Bartholow
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
199
|
Weerts EM, Wand GS, Maher B, Xu X, Stephens MA, Yang X, McCaul ME. Independent and Interactive Effects of OPRM1 and DAT1 Polymorphisms on Alcohol Consumption and Subjective Responses in Social Drinkers. Alcohol Clin Exp Res 2017; 41:1093-1104. [PMID: 28376280 DOI: 10.1111/acer.13384] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 03/27/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND The current study examined independent and interactive effects of polymorphisms of the mu opioid receptor gene (OPRM1, A118G) and variable number tandem repeats of the dopamine transporter gene (DAT1, SLC6A3) on alcohol consumption and subjective responses to alcohol in 127 young, healthy, social drinkers. METHODS Participants completed an in-person assessment, which included self-reported alcohol drinking patterns and blood sampling for DNA, and in a second visit, a cumulative alcohol dosing procedure with subjective ratings across multiple time points and breath alcohol contents (0.03 to 0.1%). DNA was analyzed for OPRM1 AA versus AG/GG (*G) genotypes, DAT1 10-repeat allele (A10) versus 9 or lesser alleles (A9), and ancestral informative markers. RESULTS There were significant epistatic interactions between OPRM1 and DAT1 genotypes. Subjective High Assessment Scale scores after alcohol consumption were highest in *G and A9 carriers, and lowest in *G and A10 carriers. Negative subjective effects were also highest in *G and A9 carriers. Effects were similar in a sensitivity analysis limited to Caucasian subjects. There were independent and epistatic interactions on drinking. The OPRM1 *G allele was independently associated with fewer heavy drinking days. The A9 allele was associated with a greater number of drinking days, which was attenuated in carriers of the *G allele. CONCLUSIONS These findings highlight the biological importance of interactions between these 2 genes and interactions between brain opioid and dopamine systems.
Collapse
Affiliation(s)
- Elise M Weerts
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gary S Wand
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Brion Maher
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Mental Health, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Xiaoqiang Xu
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mary Ann Stephens
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xiaoju Yang
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mary E McCaul
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
200
|
Wong AYL, Karppinen J, Samartzis D. Low back pain in older adults: risk factors, management options and future directions. SCOLIOSIS AND SPINAL DISORDERS 2017; 12:14. [PMID: 28435906 PMCID: PMC5395891 DOI: 10.1186/s13013-017-0121-3] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 04/04/2017] [Indexed: 12/12/2022]
Abstract
Low back pain (LBP) is one of the major disabling health conditions among older adults aged 60 years or older. While most causes of LBP among older adults are non-specific and self-limiting, seniors are prone to develop certain LBP pathologies and/or chronic LBP given their age-related physical and psychosocial changes. Unfortunately, no review has previously summarized/discussed various factors that may affect the effective LBP management among older adults. Accordingly, the objectives of the current narrative review were to comprehensively summarize common causes and risk factors (modifiable and non-modifiable) of developing severe/chronic LBP in older adults, to highlight specific issues in assessing and treating seniors with LBP, and to discuss future research directions. Existing evidence suggests that prevalence rates of severe and chronic LBP increase with older age. As compared to working-age adults, older adults are more likely to develop certain LBP pathologies (e.g., osteoporotic vertebral fractures, tumors, spinal infection, and lumbar spinal stenosis). Importantly, various age-related physical, psychological, and mental changes (e.g., spinal degeneration, comorbidities, physical inactivity, age-related changes in central pain processing, and dementia), as well as multiple risk factors (e.g., genetic, gender, and ethnicity), may affect the prognosis and management of LBP in older adults. Collectively, by understanding the impacts of various factors on the assessment and treatment of older adults with LBP, both clinicians and researchers can work toward the direction of more cost-effective and personalized LBP management for older people.
Collapse
Affiliation(s)
- Arnold YL Wong
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR China
| | - Jaro Karppinen
- Medical Research Center Oulu, Department of Physical and Rehabilitation Medicine, University of Oulu and Oulu University Hospital, Oulu, Finland
- Finnish Institute of Occupational Health, Oulu, Finland
| | - Dino Samartzis
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong, SAR China
| |
Collapse
|