1
|
Kaushik S, Ahmad F, Choudhary S, Mathkor DM, Mishra BN, Singh V, Haque S. Critical appraisal and systematic review of genes linked with cocaine addiction, depression and anxiety. Neurosci Biobehav Rev 2023; 152:105270. [PMID: 37271299 DOI: 10.1016/j.neubiorev.2023.105270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 05/13/2023] [Accepted: 06/02/2023] [Indexed: 06/06/2023]
Abstract
Recent lifestyle changes have resulted in tremendous peer pressure and mental stress, and increased the incidences of chronic psychological disorders; like addiction, depression and anxiety (ADA). In this context, the stress-tolerance levels vary amongst individuals and genetic factors play prominent roles. Vulnerable individuals may often be drawn towards drug-addiction to combat stress. This systematic review critically appraises the relationship of various genetic factors linked with the incidences of ADA development. For coherence, we focused solely on cocaine as a substance of abuse in this study. Online scholarly databases were used to screen pertinent literature using apt keywords; and the final retrieval included 42 primary-research articles. The major conclusion drawn from this systematic analysis states that there are 51 genes linked with the development of ADA; and 3 (BDNF, PERIOD2 and SLC6A4) of them are common to all the three aspects of ADA. Further, inter-connectivity analyses of the 51 genes further endorsed the central presence of BDNF and SLC6A4 genes in the development of ADA disorders. The conclusions derived from this systematic study pave the way for future studies for the identification of diagnostic biomarkers and drug targets; and for the development of novel and effective therapeutic regimens against ADA.
Collapse
Affiliation(s)
- Shradhha Kaushik
- Department of Biotechnology, Institute of Engineering and Technology, Dr. A.P.J. Abdul Kalam Technical University, Lucknow 226021, Uttar Pradesh, India
| | - Faraz Ahmad
- Department of Biotechnology, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology, Vellore 632014, India
| | - Sunita Choudhary
- Department of Biotechnology, Institute of Engineering and Technology, Dr. A.P.J. Abdul Kalam Technical University, Lucknow 226021, Uttar Pradesh, India
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Bhartendu Nath Mishra
- Department of Biotechnology, Institute of Engineering and Technology, Dr. A.P.J. Abdul Kalam Technical University, Lucknow 226021, Uttar Pradesh, India
| | - Vineeta Singh
- Department of Biotechnology, Institute of Engineering and Technology, Dr. A.P.J. Abdul Kalam Technical University, Lucknow 226021, Uttar Pradesh, India.
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon; Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, the United Arab Emirates.
| |
Collapse
|
2
|
Stress decreases serotonin tone in the nucleus accumbens in male mice to promote aversion and potentiate cocaine preference via decreased stimulation of 5-HT 1B receptors. Neuropsychopharmacology 2022; 47:891-901. [PMID: 34564712 PMCID: PMC8882182 DOI: 10.1038/s41386-021-01178-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/25/2021] [Accepted: 09/01/2021] [Indexed: 02/08/2023]
Abstract
Stress-induced release of dynorphins (Dyn) activates kappa opioid receptors (KOR) in serotonergic neurons to produce dysphoria and potentiate drug reward; however, the circuit mechanisms responsible for this effect are not known. In male mice, we found that conditional deletion of KOR from Slc6a4 (SERT)-expressing neurons blocked stress-induced potentiation of cocaine conditioned place preference (CPP). Within the dorsal raphe nucleus (DRN), two overlapping populations of KOR-expressing neurons: Slc17a8 (VGluT3) and SERT, were distinguished functionally and anatomically. Optogenetic inhibition of these SERT+ neurons potentiated subsequent cocaine CPP, whereas optical inhibition of the VGluT3+ neurons blocked subsequent cocaine CPP. SERT+/VGluT3- expressing neurons were concentrated in the lateral aspect of the DRN. SERT projections from the DRN were observed in the medial nucleus accumbens (mNAc), but VGluT3 projections were not. Optical inhibition of SERT+ neurons produced place aversion, whereas optical stimulation of SERT+ terminals in the mNAc attenuated stress-induced increases in forced swim immobility and subsequent cocaine CPP. KOR neurons projecting to mNAc were confined to the lateral aspect of the DRN, and the principal source of dynorphinergic (Pdyn) afferents in the mNAc was from local neurons. Excision of Pdyn from the mNAc blocked stress-potentiation of cocaine CPP. Prior studies suggested that stress-induced dynorphin release within the mNAc activates KOR to potentiate cocaine preference by a reduction in 5-HT tone. Consistent with this hypothesis, a transient pharmacological blockade of mNAc 5-HT1B receptors potentiated subsequent cocaine CPP. 5-HT1B is known to be expressed on 5-HT terminals in NAc, and 5-HT1B transcript was also detected in Pdyn+, Adora2a+ and ChAT+ (markers for direct pathway, indirect pathway, and cholinergic interneurons, respectively). Following stress exposure, 5-HT1B transcript was selectively elevated in Pdyn+ cells of the mNAc. These findings suggest that Dyn/KOR regulates serotonin activation of 5HT1B receptors within the mNAc and dynamically controls stress response, affect, and drug reward.
Collapse
|
3
|
Yevoo PE, Maffei A. Women in Neuroscience: Four Women’s Contributions to Science and Society. Front Integr Neurosci 2022; 15:810331. [PMID: 35153689 PMCID: PMC8825414 DOI: 10.3389/fnint.2021.810331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/27/2021] [Indexed: 11/24/2022] Open
Abstract
There has been increased cognizance of gender inequity and the importance of an inclusive and diverse approach to scientific research in recent years. However, the innovative work of women scientists is still undervalued based on reports of fewer women in leadership positions, limited citations of research spearheaded by women, reduced federal grant awards, and lack of recognition. Women have been involved in trailblazing work that paved the way for contemporary scientific inquiry. The strides made in current neuroscience include contributions from women who deserve more recognition. In this review, we discuss the work of four women whose groundbreaking scientific work has made ineffaceable marks in the neuroscience field. These women are pioneers of research and innovators and, in addition, contribute to positive change that bolsters the academic community and society. This article celebrates these women scientists, their substantial impacts in neuroscience, and the positive influence of their work on advancing society and culture.
Collapse
Affiliation(s)
- Priscilla E. Yevoo
- Department of Neurobiology and Behavior, SUNY – Stony Brook, Stony Brook, NY, United States
- Graduate Program in Neuroscience, SUNY – Stony Brook, Stony Brook, NY, United States
- *Correspondence: Priscilla E. Yevoo,
| | - Arianna Maffei
- Department of Neurobiology and Behavior, SUNY – Stony Brook, Stony Brook, NY, United States
- Graduate Program in Neuroscience, SUNY – Stony Brook, Stony Brook, NY, United States
| |
Collapse
|
4
|
Sakala K, Kasearu K, Katus U, Veidebaum T, Harro J. Association between platelet MAO activity and lifetime drug use in a longitudinal birth cohort study. Psychopharmacology (Berl) 2022; 239:327-337. [PMID: 35001146 DOI: 10.1007/s00213-021-06035-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022]
Abstract
RATIONALE Platelet monoamine oxidase (MAO) activity, a marker of central serotonergic capacity, has been associated with a variety of problem behaviours. However, studies on platelet MAO activity and addictive drugs have not consistently linked MAO activity with addiction or reported to predict illicit substance use initiation or frequency. OBJECTIVES Platelet MAO activity and illicit drug use was examined in a longitudinal birth cohort study. METHODS The sample included both birth cohorts (original n = 1238) of the Estonian Children Personality Behaviour and Health Study. Longitudinal association from age 15 to 25 years between platelet MAO activity and lifetime drug use was analysed by mixed-effects regression models. Differences at ages 15, 18 and 25 were analysed by t-test. Cox proportional hazard regression analysis was used to assess the association between platelet MAO activity and the age of drug use initiation. RESULTS Male subjects who reported at least one drug use event had lower platelet MAO activity compared to nonusers, both in cross-sectional and longitudinal analyses. Males with low platelet MAO activity had started to use drugs at a younger age. Moreover, in male subjects who had experimented with illicit drugs only once in lifetime, low platelet MAO activity was also associated with higher risk at a younger age. In females, platelet MAO activity was not associated with drug use. CONCLUSION In males, low platelet MAO activity is associated with drug abuse primarily owing to risk-taking at early age.
Collapse
Affiliation(s)
- Katre Sakala
- Department of Chronic Diseases, National Institute for Health Development, Hiiu 42, 11619, Tallinn, Estonia.,Institute of Family Medicine and Public Health, Faculty of Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia.,School of Natural Sciences and Health, Tallinn University, Narva Road 29, 10120, Tallinn, Estonia
| | - Kairi Kasearu
- Institute of Social Studies, Faculty of Social Sciences, University of Tartu, Lossi 36, 51003, Tartu, Estonia
| | - Urmeli Katus
- Institute of Family Medicine and Public Health, Faculty of Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Toomas Veidebaum
- Department of Chronic Diseases, National Institute for Health Development, Hiiu 42, 11619, Tallinn, Estonia
| | - Jaanus Harro
- School of Natural Sciences and Health, Tallinn University, Narva Road 29, 10120, Tallinn, Estonia. .,Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14a, 50411, Tartu, Estonia.
| |
Collapse
|
5
|
Abstract
The ventromedial nucleus of the hypothalamus (VMH) is a complex brain structure that is integral to many neuroendocrine functions, including glucose regulation, thermogenesis, and appetitive, social, and sexual behaviors. As such, it is of little surprise that the nucleus is under intensive investigation to decipher the mechanisms which underlie these diverse roles. Developments in genetic and investigative tools, for example the targeting of steroidogenic factor-1-expressing neurons, have allowed us to take a closer look at the VMH, its connections, and how it affects competing behaviors. In the current review, we aim to integrate recent findings into the literature and contemplate the conclusions that can be drawn.
Collapse
Affiliation(s)
- Tansi Khodai
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, UK
| | - Simon M Luckman
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, UK
- Correspondence: Simon M. Luckman, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, UK.
| |
Collapse
|
6
|
Muderrisoglu A, Babaoglu E, Korkmaz ET, Ongun MC, Karabulut E, Iskit AB, Emri S, Babaoglu MO. Effects of Genetic Polymorphisms of Drug Transporter ABCB1 (MDR1) and Cytochrome P450 Enzymes CYP2A6, CYP2B6 on Nicotine Addiction and Smoking Cessation. Front Genet 2020; 11:571997. [PMID: 33329709 PMCID: PMC7734344 DOI: 10.3389/fgene.2020.571997] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/12/2020] [Indexed: 01/08/2023] Open
Abstract
Objectives To determine the effects of genetic polymorphisms of ABCB1 (MDR1), CYP2A6, CYP2B6 on smoking status, and clinical outcomes of smoking cessation therapies in a Turkish population. Methods 130 smokers and 130 non-smokers were recruited. Individuals who never smoked were described as non-smokers. 130 smokers were treated with nicotine replacement therapy (NRT) (n = 40), bupropion (n = 47), bupropion + NRT (n = 15), and varenicline (n = 28). Smokers were checked by phone after 12 weeks of treatment whether they were able to quit smoking or not. Genotyping and phenotyping were performed. Results Cessation rates were as follows; 20.0% for NRT, 29.8% for bupropion, 40.0% for bupropion + NRT, 57.1% for varenicline (p = 0.013). The frequency of ABCB1 1236TT-2677TT-3435TT haplotype was significantly higher in non-smokers as compared to smokers (21.5% vs. 10.8, respectively; p = 0.018). Neither smoking status nor smoking cessation rates were associated with genetic variants of CYP2A6 (p = 0.652, p = 0.328, respectively), or variants of CYP2B6 (p = 0.514, p = 0.779, respectively). Conclusion Genetic variants of the drug transporter ABCB1 and the 1236TT-2677TT-3435TT haplotype was significantly associated with non-smoking status. Neither ABCB1 nor CYP2A6, CYP2B6 genetic variants were associated with smoking cessation rates at the 12th week of drug treatment.
Collapse
Affiliation(s)
- Ahmet Muderrisoglu
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Elif Babaoglu
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Elif Tugce Korkmaz
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Mert C Ongun
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Erdem Karabulut
- Department of Biostatistics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Alper B Iskit
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Salih Emri
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Melih O Babaoglu
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
7
|
Randesi M, Levran O, van den Brink W, Blanken P, van Ree JM, Ott J, Kreek MJ. Further evidence for the association of GAL, GALR1 and NPY1R variants with opioid dependence. Pharmacogenomics 2020; 21:903-917. [PMID: 32757697 PMCID: PMC7487975 DOI: 10.2217/pgs-2020-0045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: Heroin addiction is a chronic, relapsing disease that has genetic and environmental, including drug-induced, contributions. Stress influences the development of addictions. This study was conducted to determine if variants in stress-related genes are associated with opioid dependence (OD). Patients & methods: One hundred and twenty variants in 26 genes were analyzed in 597 Dutch subjects. Patients included 281 OD in methadone maintenance with or without heroin-assisted treatment and 316 controls. Results: Twelve SNPs in seven genes showed a nominally significant association with OD. Experiment-wise significant associations (p < 0.05) were found for three SNP pairs, through an interaction effect: NPY1R/GAL rs4691910/rs1893679, NPY1R/GAL rs4691910/rs3136541 and GALR1/GAL rs9807208/rs3136541. Conclusion: This study lends more evidence to previous reports of association of stress-related variants with heroin dependence.
Collapse
Affiliation(s)
- Matthew Randesi
- Laboratory of The Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA,Author for correspondence:
| | - Orna Levran
- Laboratory of The Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Wim van den Brink
- Department of Psychiatry, Amsterdam Institute for Addiction Research, Academic Medical Center, University of Amsterdam, PO Box 22660, 1100DD Amsterdam, The Netherlands
| | - Peter Blanken
- Parnassia Addiction Research Centre (Brijder Addiction Treatment), PO Box 53002, 2505 AA The Hague, The Netherlands
| | - Jan M van Ree
- Brain Center Rudolf Magnus, University Medical Center, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Jurg Ott
- Laboratory of Statistical Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Mary J Kreek
- Laboratory of The Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| |
Collapse
|
8
|
Sandoval-Sierra JV, Salgado García FI, Brooks JH, Derefinko KJ, Mozhui K. Effect of short-term prescription opioids on DNA methylation of the OPRM1 promoter. Clin Epigenetics 2020; 12:76. [PMID: 32493461 PMCID: PMC7268244 DOI: 10.1186/s13148-020-00868-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022] Open
Abstract
Background A long-term opioid use has been associated with hypermethylation of the opioid receptor mu 1 (OPRM1) promoter. Very little is currently known about the early epigenetic response to therapeutic opioids. Here, we examine whether we can detect DNA methylation changes associated with a few days’ use of prescribed opioids. Genome-wide DNA methylation was assayed in a cohort of 33 opioid-naïve participants who underwent standard dental surgery followed by opioid self-administration. Saliva samples were collected before surgery (visit 1), and at two postsurgery visits at 2.7 ± 1.5 days (visit 2), and 39 ± 10 days (visit 3) after the discontinuation of opioid analgesics. Results The perioperative methylome underwent significant changes over the three visits that were primarily due to postoperative inflammatory response and cell heterogeneity. To specifically examine the effect of opioids, we started with a candidate gene approach and evaluated 10 CpGs located in the OPRM1 promoter. There was a significant cross-sectional variability in opioid use, and for participants who self-administered the prescribed drugs, the total dosage ranged from 5–210 morphine milligram equivalent (MME). Participants were categorized by cumulative dosage into three groups: < 25 MME, 25–90 MME, and ≥ 90 MME. Using mixed-effects modeling, 4 CpGs had significant positive associations with opioid dose at two-tailed p value < 0.05, and overall, 9 of the 10 OPRM1 promoter CpGs showed the predicted higher methylation in the higher dose groups relative to the lowest dose group. After adjustment for age, cellular heterogeneity, and past tobacco use, the promoter mean methylation also had positive associations with cumulative MME (regression coefficient = 0.0002, one-tailed p value = 0.02) and duration of opioid use (regression coefficient = 0.003, one-tailed p value = 0.001), but this effect was significant only for visit 3. A preliminary epigenome-wide association study identified a significant CpG in the promoter of the RAS-related signaling gene, RASL10A, that may be predictive of opioid dosage. Conclusion The present study provides evidence that the hypermethylation of the OPRM1 promoter is in response to opioid use and that epigenetic differences in OPRM1 and other sites are associated with a short-term use of therapeutic opioids.
Collapse
Affiliation(s)
- Jose Vladimir Sandoval-Sierra
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Francisco I Salgado García
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jeffrey H Brooks
- Department of Oral and Maxillofacial Surgery, College of Dentistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Karen J Derefinko
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Khyobeni Mozhui
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| |
Collapse
|
9
|
Moses TE, Burmeister M, Greenwald MK. Heroin delay discounting and impulsivity: Modulation by DRD1 genetic variation. Addict Biol 2020; 25:e12777. [PMID: 31192519 DOI: 10.1111/adb.12777] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/11/2019] [Accepted: 04/27/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dopamine D1 receptors (encoded by DRD1) are implicated in drug addiction and high-risk behaviors. Delay discounting (DD) procedures measure decisional balance between choosing smaller/sooner rewards vs larger/later rewards. Individuals with higher DD (rapid discounting) are prone to maladaptive behaviors that provide immediate reinforcement (eg, substance use). DRD1 variants have been linked with increased DD (in healthy volunteers) and opioid abuse. This study determined whether four dopaminergic functional variants modulated heroin DD and impulsivity. METHODS Substance use, DD, and genotype data (DRD1 rs686 and rs5326, DRD3 rs6280, COMT rs4680) were obtained from 106 current heroin users. Subjects completed an array of DD choices during two imagined conditions: heroin satiation and withdrawal. Rewards were expressed as $10 heroin bag units, with maximum delayed amount of 30 bags. Delays progressively increased from 3 to 96 hours. RESULTS DRD1 rs686 (A/A, n = 25; G/A, n = 56; G/G, n = 25) was linearly related to the difference in heroin DD (area under the curve; AUC) between the heroin satiation and withdrawal conditions; specifically, G/G homozygotes had a significantly smaller (satiation minus withdrawal) AUC difference score had higher drug-use impulsivity questionnaire scores, relative to A/A homozygotes, with G/A intermediate. DRD3 and COMT variants were not associated with these DD and impulsivity outcomes. CONCLUSION DRD1 rs686 modulated the difference in heroin DD score between pharmacological states and was associated with drug-use impulsivity. These data support a role of DRD1 in opioid DD and impulsive behaviors.
Collapse
|
10
|
Koijam AS, Chakraborty B, Mukhopadhyay K, Rajamma U, Haobam R. A single nucleotide polymorphism in OPRM1(rs483481) and risk for heroin use disorder. J Addict Dis 2020; 38:214-222. [PMID: 32189578 DOI: 10.1080/10550887.2020.1740070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Opioid receptor mu1 (OPRM1) is the target of many opioid drugs, and it is known to have affinity toward both endogenous and exogenous opioids, opiate and opioid analgesic drugs. The present study was undertaken to explore association of single nucleotide polymorphisms (SNPs) in the OPRM1 gene with heroin use disorder. Ten OPRM1 polymorphisms were analyzed in 132 cases and 147 healthy controls. The SNP rs483481 showed significant allelic, genotypic and haplotypic association (Allelic: p-value = 0.003, OR = 1.75, CI = 1.21-2.55) (Genotypic: p-value = 0.003, OR = 1.72, CI = 1.08-2.75) with heroin use disorder. Allelic and genotypic association remained significant even after multiple testing corrections with 1000 permutations. A significant positive correlation between 'Number of times drug abstained' and 'rs483481-AA genotype' (p-value = 0.002; Pearson correlation = 0.265) was also observed. One-way ANOVA analysis demonstrated significant association of rs483481 with 'number of times drug abstained' (F = 4.86, p-value =0.009). 'A' allele and 'AA' genotype of marker rs483481 seem to confer protective effect while 'G' allele and 'GG' genotype potentiates risk for heroin use disorder. OPRM1 is found to be associated with heroin use disorder in the studied Manipuri cohort. The study suggests that individuals with G allele and GG genotypes at rs483481 could be more vulnerable to heroin dependence, and it could be taken into consideration in prevention and intervention programs.
Collapse
Affiliation(s)
| | - Barnali Chakraborty
- Manovikas Biomedical Research & Diagnostic Centre, Manovikas Kendra, Kolkata, West Bengal, India
| | - Kanchan Mukhopadhyay
- Manovikas Biomedical Research & Diagnostic Centre, Manovikas Kendra, Kolkata, West Bengal, India
| | - Usha Rajamma
- Manovikas Biomedical Research & Diagnostic Centre, Manovikas Kendra, Kolkata, West Bengal, India.,Centre for Development & Aging Research, Inter University Centre for Biomedical Research & Super Speciality Hospital (IUCBR&SSH), MG University Campus at Thalappady, Kottayam, Kerala, India
| | - Reena Haobam
- Department of Biotechnology, Manipur University, Imphal, Manipur, India
| |
Collapse
|
11
|
The association between the OPRM1 A118G polymorphism and addiction in a Turkish population. Arh Hig Rada Toksikol 2020; 70:97-103. [PMID: 31246565 DOI: 10.2478/aiht-2019-70-3153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 04/01/2019] [Indexed: 01/12/2023] Open
Abstract
Susceptibility to addiction has a complex genetic basis that includes genes associated with the action and metabolism of drugs of abuse. One important gene in that respect is OPRM1, which codes for the μ-opioid receptor and has an important role in mediating the rewarding effects of addiction substances. The aim of our study was to assess the prevalence of the OPRM1 A118G polymorphism (rs1799971) in Turkish population and to investigate its association with opioid and other substance addiction. In addition, we examined the association of rs1799971 in addicted patients who were also diagnosed with psychiatric disorders. The study included 103 patients addicted to opioids, cocaine, ecstasy, alcohol, lysergic acid diethylamide (LSD), cannabis, and sedative/hypnotic substances and 83 healthy volunteers with similar demographic features as controls. rs1799971 polymorphisms were identified with the polymerase chain reaction restriction fragment length polymorphism method (PCR-RFLP). The genotype frequencies were significantly higher in the addicted patients than controls (32.0 % vs 16.9 %, respectively; p=0.027). The prevalence of the G allele was 16.1 % in the addicted group and 8.4 % in the control group (p=0.031). Our study confirmed the association between the rs1799971(G) allele frequency and opioid and other substance addiction, but not with psychiatric disorders.
Collapse
|
12
|
Zortea M, Ramalho L, Alves RL, Alves CFDS, Braulio G, Torres ILDS, Fregni F, Caumo W. Transcranial Direct Current Stimulation to Improve the Dysfunction of Descending Pain Modulatory System Related to Opioids in Chronic Non-cancer Pain: An Integrative Review of Neurobiology and Meta-Analysis. Front Neurosci 2019; 13:1218. [PMID: 31803005 PMCID: PMC6876542 DOI: 10.3389/fnins.2019.01218] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/29/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Opioid long-term therapy can produce tolerance, opioid-induced hyperalgesia (OIH), and it induces dysfunction in pain descending pain inhibitory system (DPIS). Objectives: This integrative review with meta-analysis aimed: (i) To discuss the potential mechanisms involved in analgesic tolerance and opioid-induced hyperalgesia (OIH). (ii) To examine how the opioid can affect the function of DPIS. (ii) To show evidence about the tDCS as an approach to treat acute and chronic pain. (iii) To discuss the effect of tDCS on DPIS and how it can counter-regulate the OIH. (iv) To draw perspectives for the future about the tDCS effects as an approach to improve the dysfunction in the DPIS in chronic non-cancer pain. Methods: Relevant published randomized clinical trials (RCT) comparing active (irrespective of the stimulation protocol) to sham tDCS for treating chronic non-cancer pain were identified, and risk of bias was assessed. We searched trials in PubMed, EMBASE and Cochrane trials databases. tDCS protocols accepted were application in areas of the primary motor cortex (M1), dorsolateral prefrontal cortex (DLPFC), or occipital area. Results: Fifty-nine studies were fully reviewed, and 24 with moderate to the high-quality methodology were included. tDCS improved chronic pain with a moderate effect size [pooled standardized mean difference; -0.66; 95% confidence interval (CI) -0.91 to -0.41]. On average, active protocols led to 27.26% less pain at the end of treatment compared to sham [95% CI; 15.89-32.90%]. Protocol varied in terms of anodal or cathodal stimulation, areas of stimulation (M1 and DLPFC the most common), number of sessions (from 5 to 20) and current intensity (from 1 to 2 mA). The time of application was 20 min in 92% of protocols. Conclusion: In comparison with sham stimulation, tDCS demonstrated a superior effect in reducing chronic pain conditions. They give perspectives that the top-down neuromodulator effects of tDCS are a promising approach to improve management in refractory chronic not-cancer related pain and to enhance dysfunctional neuronal circuitries involved in the DPIS and other pain dimensions and improve pain control with a therapeutic opioid-free. However, further studies are needed to determine individualized protocols according to a biopsychosocial perspective.
Collapse
Affiliation(s)
- Maxciel Zortea
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Leticia Ramalho
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Rael Lopes Alves
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Camila Fernanda da Silveira Alves
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Gilberto Braulio
- Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.,Service of Anesthesia and Perioperative Medicine, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Iraci Lucena da Silva Torres
- Department of Pharmacology, Institute of Health Sciences (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Pharmacology of Pain and Neuromodulation: Pre-clinical Investigations Research Group, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil
| | - Felipe Fregni
- Neuromodulation Center, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, United States
| | - Wolnei Caumo
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.,Pain Treatment and Palliative Medicine Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| |
Collapse
|
13
|
Moses TEH, Greenwald MK. History of regular nonmedical sedative and/or alcohol use differentiates substance-use patterns and consequences among chronic heroin users. Addict Behav 2019; 97:14-19. [PMID: 31112911 PMCID: PMC6581601 DOI: 10.1016/j.addbeh.2019.05.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/15/2019] [Accepted: 05/15/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Concurrent use of sedating substances (e.g. alcohol or benzodiazepines) with opioids is associated with increased negative consequences of opioid use; however, few studies have attempted to differentiate effects of using sedating substances on heroin-use outcomes. This study examines differences between heroin users who use alcohol or misuse sedatives regularly and those who do not. METHODS Substance-use data were collected from 367 non-treatment seeking, chronic heroin-using, 18-to-55 year-old participants. We created 4 groups based on self-reported lifetime history of regular (at least weekly) substance use: heroin only (n = 95), heroin and sedatives (n = 21), heroin and alcohol (n = 151), and heroin, sedative, and alcohol (n = 100). Chi-square analyses and ANOVAs with Bonferroni post hoc tests were used to explore differences between these groups. RESULTS Heroin users who denied lifetime alcohol or nonmedical sedative use regularly endorsed fewer consequences associated with any substance they had used. Total adverse consequences of heroin use (e.g. health problems) were significantly higher among those who misused sedatives regularly, irrespective of alcohol use history (F(3,361) = 10.21; p < .001). Regular alcohol use did not independently impact heroin consequences but was associated with increased use of other substances. CONCLUSIONS Although polysubstance use is normative among heroin users, the risks depend on the substances used. Regular sedative use is associated with increased heroin consequences whereas regular alcohol use is not. This study refines the investigation of polysubstance use and highlights subgroup differences depending on types of substances used regularly. This knowledge is critical for understanding substance-use motivations and creating avenues for harm reduction.
Collapse
Affiliation(s)
- Tabitha E H Moses
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Mark K Greenwald
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
14
|
Wang CW, Ma M, Lu WG, Luo RQ. Association between prodynorphin gene polymorphisms and opioid dependence susceptibility: a meta-analysis. BMC Psychiatry 2019; 19:281. [PMID: 31510971 PMCID: PMC6737717 DOI: 10.1186/s12888-019-2272-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 09/04/2019] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Prodynorphin (PDYN) gene polymorphisms have been linked with opioid dependence (OD) with conflicting outcomes, the aim of this study is to synthesize the existing evidence of the association between PDYN polymorphisms and OD susceptibility. METHODS Four databases including PubMed, EMBASE, Web of Science, and Wanfang were retrieved for relevant studies before August, 2018. All identified studies were evaluated using predetermined inclusion and exclusion criteria. Summary odds ratio (OR) and 95% confidence interval (95%CI) were calculated to appraise the association. Statistical analysis was performed using RevMan 5.3 software. RESULTS A total of seven case-control studies with 3129 cases and 3289 controls were recruited in the meta-analysis. For rs910080, rs1997794, rs1022563, and rs2235749 polymorphisms of PDYN gene, there were six, four, five, and four studies eventually included, respectively. The findings indicated that rs910080 polymorphism was significantly correlated with OD among Asian population under allelic model (A vs. G, OR = 1.30, 95% CI 1.04-1.62, P = 0.02, FDR = 0.05) and dominant model (AA+AG vs. GG, OR = 1.25, 95% CI 1.04-1.51, P = 0.02, FDR = 0.05). However, rs1022563, rs1997794 and rs2235749 polymorphisms did not appear to associate with OD susceptibility. CONCLUSIONS There existed a significant association between rs1022563 polymorphism and OD among Asian population. As the included studies were not adequate to guarantee a robust and convincing conclusion, future studies with larger sample size among more ethnicities are recommended.
Collapse
Affiliation(s)
- Chang-wang Wang
- Department of Psychiatry, Wuchang Hospital, South Luoshi Avenue 505#, Hongshan District, Wuhan, 430070 China
| | - Min Ma
- grid.413247.7Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Wei-guang Lu
- Department of Psychiatry, Wuchang Hospital, South Luoshi Avenue 505#, Hongshan District, Wuhan, 430070 China
| | - Ru-qin Luo
- Department of Psychiatry, Wuchang Hospital, South Luoshi Avenue 505#, Hongshan District, Wuhan, 430070 China
| |
Collapse
|
15
|
Awad ME, Padela MT, Sayeed Z, El-Othmani MM, Zekaj M, Darwiche HF, Saleh KJ. Pharmacogenomic Testing for Postoperative Pain Optimization Before Total Joint Arthroplasty: A Focus on Drug-Drug-Gene Interaction with Commonly Prescribed Drugs and Prior Opioid Use. JBJS Rev 2019; 7:e2. [PMID: 31094889 DOI: 10.2106/jbjs.rvw.18.00058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Mohamed E Awad
- Resident Research Partnership, Detroit, Michigan.,FAJR Scientific, Detroit, Michigan.,Michigan State University College of Osteopathic Medicine, Detroit, Michigan.,John D. Dingell VA Medical Center, Detroit, Michigan
| | - Muhammad Talha Padela
- Resident Research Partnership, Detroit, Michigan.,FAJR Scientific, Detroit, Michigan.,John D. Dingell VA Medical Center, Detroit, Michigan.,Department of Orthopaedic Surgery and Sports Medicine, Detroit Medical Center, Detroit, Michigan.,Department of Orthopaedic Surgery, Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois
| | - Zain Sayeed
- Resident Research Partnership, Detroit, Michigan.,John D. Dingell VA Medical Center, Detroit, Michigan.,Department of Orthopaedic Surgery and Sports Medicine, Detroit Medical Center, Detroit, Michigan.,Department of Orthopaedic Surgery, Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois
| | - Mouhanad M El-Othmani
- Department of Orthopaedic Surgery and Sports Medicine, Detroit Medical Center, Detroit, Michigan
| | - Mark Zekaj
- Department of Orthopaedic Surgery and Sports Medicine, Detroit Medical Center, Detroit, Michigan
| | - Hussein F Darwiche
- Department of Orthopaedic Surgery and Sports Medicine, Detroit Medical Center, Detroit, Michigan
| | - Khaled J Saleh
- FAJR Scientific, Detroit, Michigan.,Michigan State University College of Osteopathic Medicine, Detroit, Michigan.,John D. Dingell VA Medical Center, Detroit, Michigan
| |
Collapse
|
16
|
Burns JA, Kroll DS, Feldman DE, Kure Liu C, Manza P, Wiers CE, Volkow ND, Wang GJ. Molecular Imaging of Opioid and Dopamine Systems: Insights Into the Pharmacogenetics of Opioid Use Disorders. Front Psychiatry 2019; 10:626. [PMID: 31620026 PMCID: PMC6759955 DOI: 10.3389/fpsyt.2019.00626] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/05/2019] [Indexed: 12/21/2022] Open
Abstract
Opioid use in the United States has steadily risen since the 1990s, along with staggering increases in addiction and overdose fatalities. With this surge in prescription and illicit opioid abuse, it is paramount to understand the genetic risk factors and neuropsychological effects of opioid use disorder (OUD). Polymorphisms disrupting the opioid and dopamine systems have been associated with increased risk for developing substance use disorders. Molecular imaging studies have revealed how these polymorphisms impact the brain and contribute to cognitive and behavioral differences across individuals. Here, we review the current molecular imaging literature to assess how genetic variations in the opioid and dopamine systems affect function in the brain's reward, cognition, and stress pathways, potentially resulting in vulnerabilities to OUD. Continued research of the functional consequences of genetic variants and corresponding alterations in neural mechanisms will inform prevention and treatment of OUD.
Collapse
Affiliation(s)
- Jamie A Burns
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Danielle S Kroll
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Dana E Feldman
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | | | - Peter Manza
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Corinde E Wiers
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States.,National Institute on Drug Abuse, Bethesda, MD, United States
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| |
Collapse
|
17
|
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
|
18
|
Hashemi M, Shakiba M, Sanaei S, Shahkar G, Rezaei M, Mojahed A, Bahari G. Evaluation of prodynorphin gene polymorphisms and their association with heroin addiction in a sample of the southeast Iranian population. MOLECULAR BIOLOGY RESEARCH COMMUNICATIONS 2018; 7:1-6. [PMID: 29911117 PMCID: PMC5991531 DOI: 10.22099/mbrc.2017.27182.1294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Genetic factors are supposed to account for about 30-50% of the predisposition to cocaine and heroin addiction. This study aims at investigating the effect of rs2281285, rs2235749, rs910080 and 68bp VNTR polymorphisms of prodynorphin (PDYN) gene on heroin dependence risk in a sample of the southeast Iranian population. This case-control study was done on 216 heroin dependence subjects and 219 healthy subjects. Genomic DNA was extracted from peripheral blood cells using salting out method. Genotyping of PDYN polymorphisms were performed using polymerase chain reaction (PCR) or PCR-RFLP method. The findings showed that PDYN rs910080 T>C variant significantly increased the risk of heroin dependence (OR=7.91, 95%CI=3.36-18.61, P<0.0001, CC vs TT; OR=7.53, 95%CI=3.30-17.16, P<0.0001, CC vs TT+TC; OR=1.75, 95%CI=1.33-2.32, p<0.0001, C vs T). The rs2235749 C>T, rs2281285 A>G and 68bp VNTR variants of PDYN gene were not associated with heroin dependence. Altogether, our results provide an association between rs910080 polymorphism of PDYN gene and risk of heroin dependence in a sample of the southeast Iranian population.
Collapse
Affiliation(s)
- Mohammad Hashemi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mansour Shakiba
- Department of Psychiatry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Sara Sanaei
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ghazaleh Shahkar
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Maryam Rezaei
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Azizolla Mojahed
- Department of Clinical Psychology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Gholamreza Bahari
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
19
|
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
|
20
|
Wendt FR, Sajantila A, Budowle B. Predicted activity of UGT2B7, ABCB1, OPRM1, and COMT using full-gene haplotypes and their association with the CYP2D6-inferred metabolizer phenotype. Forensic Sci Int Genet 2018; 33:48-58. [DOI: 10.1016/j.fsigen.2017.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/01/2017] [Accepted: 11/22/2017] [Indexed: 12/20/2022]
|
21
|
Mongi-Bragato B, Avalos MP, Guzmán AS, Bollati FA, Cancela LM. Enkephalin as a Pivotal Player in Neuroadaptations Related to Psychostimulant Addiction. Front Psychiatry 2018; 9:222. [PMID: 29892236 PMCID: PMC5985699 DOI: 10.3389/fpsyt.2018.00222] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 05/08/2018] [Indexed: 12/21/2022] Open
Abstract
Enkephalin expression is high in mesocorticolimbic areas associated with psychostimulant-induced behavioral and neurobiological effects, and may also modulate local neurotransmission in this circuit network. Psychostimulant drugs, like amphetamine and cocaine, significantly increase the content of enkephalin in these brain structures, but we do not yet understand the specific significance of this drug-induced adaptation. In this review, we summarize the neurochemical and molecular mechanism of psychostimulant-induced enkephalin activation in mesocorticolimbic brain areas, and the contribution of this opioid peptide in the pivotal neuroadaptations and long-term behavioral changes underlying psychostimulant addiction. There is evidence suggesting that adaptive changes in enkephalin content in the mesocorticolimbic circuit, induced by acute and chronic psychostimulant administration, may represent a key initial step in the long-term behavioral and neuronal plasticity induced by these drugs.
Collapse
Affiliation(s)
- Bethania Mongi-Bragato
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María P Avalos
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrea S Guzmán
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Flavia A Bollati
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Liliana M Cancela
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
22
|
Risk of Opioid Abuse and Biopsychosocial Characteristics Associated With This Risk Among Chronic Pain Patients Attending a Multidisciplinary Pain Treatment Facility. Clin J Pain 2017; 32:859-69. [PMID: 26670615 DOI: 10.1097/ajp.0000000000000337] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The objectives of this study were to (1) determine the proportion of patients referred to a multidisciplinary pain treatment facility at risk of opioid abuse, (2) examine biopsychosocial factors associated with this risk, and (3) compare patient outcomes 6 months later across risk of opioid abuse and type of treatment (opioids vs. no opioids). METHODS Participants were 3040 patients (mean age=53.3±14.7 y; female=56%) enrolled in the Quebec Pain Registry between July 2012 and May 2014. Patients answered self-report and nurse-administered questionnaires (pain and psychosocial constructs, Opioid Risk Tool, pain medication, etc.) before initiating treatment at the multidisciplinary pain treatment facility and 6 months later. Data were analyzed using the Pearson χ tests, multivariable binary logistic regression, and multivariate general linear model. RESULTS Results showed that 81%, 13%, and 6% of patients were at low, moderate, and severe risk of opioid abuse, respectively. Civil status, pain duration, mental health-related quality of life, and cigarette smoking were significantly associated with risk of opioid abuse (P<0.001). There was a significant interaction between risk of opioid abuse and type of treatment in predicting 6-month pain outcomes and quality of life. DISCUSSION Almost 20% of patients had a moderate/severe risk of opioid abuse; whether these patients were taking opioids or not for their pain, they had worse outcomes at follow-up. These results point to the importance of assessing risk of opioid abuse in chronic pain patients and to consider how this risk may impact on their clinical evolution.
Collapse
|
23
|
Patriquin MA, Hamon SC, Harding MJ, Nielsen EM, Newton TF, De La Garza R, Nielsen DA. Genetic moderation of cocaine subjective effects by variation in the TPH1, TPH2, and SLC6A4 serotonin genes. Psychiatr Genet 2017; 27:178-186. [PMID: 28590957 PMCID: PMC5572746 DOI: 10.1097/ypg.0000000000000178] [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: 12/12/2022]
Abstract
OBJECTIVE This study investigated variants of tryptophan hydroxylase (TPH)1, TPH2, and SLC6A4 in the moderation of the subjective effects of cocaine. METHODS Non-treatment-seeking cocaine-dependent individuals (N=66) were intravenously administered saline and cocaine (40 mg) in a randomized order. Participants self-reported subjective effects of cocaine using a visual analog scale starting before administration of saline or cocaine (-15 min) to up to 20 min after infusion. Self-report ratings on the visual analog scale ranged from 0 (no effect) to 100 (greatest effect). Participants were genotyped for the TPH1 rs1799913, TPH2 rs4290270, and SLC6A4 5-HTTLPR variants. Repeated-measures analysis of covariance was used to examine changes in subjective effect scores over time while controlling for population structure. RESULTS Participants carrying the TPH1 rs1799913 A allele reported greater subjective response to cocaine for 'stimulated' and 'access' relative to the CC genotype group. Those carrying the TPH2 rs4290270 A allele reported higher 'good effect' and lower 'depressed' effect relative to the TT genotype group. Those carrying the SLC6A4 5-HTTLPR S' allele reported greater 'desire' and 'access' compared with the L'L' genotype group. CONCLUSION These findings indicate that TPH1, TPH2, and SLC6A4 variants moderate the subjective effects of cocaine in non-treatment-seeking cocaine-dependent participants.
Collapse
Affiliation(s)
- Michelle A. Patriquin
- The Menninger Clinic
- The Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
| | - Sara C. Hamon
- Statistical and Genetic Consulting LLC, Daren, CT USA
| | - Mark J. Harding
- The Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
- Michael E. DeBakey Veterans Affairs Medical Center; Houston, TX USA
| | - Ellen M. Nielsen
- The Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
- Michael E. DeBakey Veterans Affairs Medical Center; Houston, TX USA
| | - Thomas F. Newton
- The Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
| | - Richard De La Garza
- The Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
- Michael E. DeBakey Veterans Affairs Medical Center; Houston, TX USA
| | - David A. Nielsen
- The Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
- Michael E. DeBakey Veterans Affairs Medical Center; Houston, TX USA
| |
Collapse
|
24
|
Marie-Claire C, Jourdaine C, Lépine JP, Bellivier F, Bloch V, Vorspan F. Pharmacoepigenomics of opiates and methadone maintenance treatment: current data and perspectives. Pharmacogenomics 2017; 18:1359-1372. [DOI: 10.2217/pgs-2017-0040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Current treatments of opioid addiction include primarily maintenance medications such as methadone. Chronic exposure to opiate and/or long-lasting maintenance treatment induce modulations of gene expression in brain and peripheral tissues. There is increasing evidence that epigenetic modifications underlie these modulations. This review summarizes published results on opioid-induced epigenetic changes in animal models and in patients. The epigenetic modifications observed with other drugs of abuse often used by opiate abusers are also outlined. Specific methadone maintenance treatment induced epigenetic modifications at different treatment stages may be combined with the ones resulting from patients’ substance use history. Therefore, research comparing groups of addicts with similar history and substances use disorders but contrasting for well-characterized treatment phenotypes should be encouraged.
Collapse
Affiliation(s)
- Cynthia Marie-Claire
- Variabilité de réponse aux psychotropes, INSERMU1144/Faculté de Pharmacie de Paris/Université Paris Descartes/Université ParisDiderot/Université Sorbonne Paris Cité, Paris, France
| | - Clément Jourdaine
- AP-HP, GH Saint-Louis – Lariboisière – F. Widal, Pôle de Psychiatrie et de Médecine Addictologique, 75475 Paris cedex 10, France
| | - Jean-Pierre Lépine
- AP-HP, GH Saint-Louis – Lariboisière – F. Widal, Pôle de Psychiatrie et de Médecine Addictologique, 75475 Paris cedex 10, France
| | - Frank Bellivier
- Variabilité de réponse aux psychotropes, INSERMU1144/Faculté de Pharmacie de Paris/Université Paris Descartes/Université ParisDiderot/Université Sorbonne Paris Cité, Paris, France
- AP-HP, GH Saint-Louis – Lariboisière – F. Widal, Pôle de Psychiatrie et de Médecine Addictologique, 75475 Paris cedex 10, France
| | - Vanessa Bloch
- Variabilité de réponse aux psychotropes, INSERMU1144/Faculté de Pharmacie de Paris/Université Paris Descartes/Université ParisDiderot/Université Sorbonne Paris Cité, Paris, France
| | - Florence Vorspan
- Variabilité de réponse aux psychotropes, INSERMU1144/Faculté de Pharmacie de Paris/Université Paris Descartes/Université ParisDiderot/Université Sorbonne Paris Cité, Paris, France
- AP-HP, GH Saint-Louis – Lariboisière – F. Widal, Pôle de Psychiatrie et de Médecine Addictologique, 75475 Paris cedex 10, France
| |
Collapse
|
25
|
Wendt FR, Pathak G, Sajantila A, Chakraborty R, Budowle B. Global genetic variation of select opiate metabolism genes in self-reported healthy individuals. THE PHARMACOGENOMICS JOURNAL 2017; 18:281-294. [PMID: 28398354 DOI: 10.1038/tpj.2017.13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 02/16/2017] [Accepted: 02/21/2017] [Indexed: 12/26/2022]
Abstract
CYP2D6 is a key pharmacogene encoding an enzyme impacting poor, intermediate, extensive and ultrarapid phase I metabolism of many marketed drugs. The pharmacogenetics of opiate drug metabolism is particularly interesting due to the relatively high incidence of addiction and overdose. Recently, trans-acting opiate metabolism and analgesic response enzymes (UGT2B7, ABCB1, OPRM1 and COMT) have been incorporated into pharmacogenetic studies to generate more comprehensive metabolic profiles of patients. With use of massively parallel sequencing, it is possible to identify additional polymorphisms that fine tune, or redefine, previous pharmacogenetic findings, which typically rely on targeted approaches. The 1000 Genomes Project data were analyzed to describe population genetic variation and statistics for these five genes in self-reported healthy individuals in five global super- and 26 sub-populations. Findings on the variation of these genes in various populations expand baseline understanding of pharmacogenetically relevant polymorphisms for future studies of affected cohorts.
Collapse
Affiliation(s)
- F R Wendt
- Institute for Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - G Pathak
- Institute for Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - A Sajantila
- Department of Forensic Medicine, University of Helsinki, Helsinki, Finland
| | - R Chakraborty
- Institute for Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - B Budowle
- Institute for Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA.,Center for Human Identification, University of North Texas Health Science Center, Fort Worth, TX USA.,Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
26
|
Biochemical Diagnosis in Substance and Non-substance Addiction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1010:169-202. [PMID: 29098673 DOI: 10.1007/978-981-10-5562-1_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
An optimal biochemical marker for addiction would be some easily traced molecules in body specimens, which indicates indulgent addictive behaviors, or susceptibility to certain addictive stimuli. In this chapter, we discussed existing literature about possible biomarkers, and classified them into three categories: origin forms and metabolites of substances, markers from biochemical responses to certain addiction, and genetic and epigenetic biomarkers suggesting susceptibility to addiction. In every category, we examined studies concerning certain type of addiction one by one, with focuses mainly on opiates, psychostimulants, and pathological gambling. Several promising molecules were highlighted, including those of neurotrophic factors, inflammatory factors, and indicators of vascular injury, and genetic and epigenetic biomarkers such as serum miRNAs. DNA methylation signatures and signal nucleotide polymorphism of candidate gene underlying the addiction.
Collapse
|
27
|
Crist RC, Doyle GA, Nelson EC, Degenhardt L, Martin NG, Montgomery GW, Saxon AJ, Ling W, Berrettini WH. A polymorphism in the OPRM1 3'-untranslated region is associated with methadone efficacy in treating opioid dependence. THE PHARMACOGENOMICS JOURNAL 2016; 18:173-179. [PMID: 27958381 PMCID: PMC5468510 DOI: 10.1038/tpj.2016.89] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/07/2016] [Accepted: 11/14/2016] [Indexed: 11/10/2022]
Abstract
The mu-opioid receptor (MOR) is the primary target of methadone and buprenorphine. The primary neuronal transcript of the OPRM1 gene, MOR-1, contains a ~13kb 3′ untranslated region with five common haplotypes in European-Americans. We analyzed the effects of these haplotypes on the percentage of opioid positive urine tests in European-Americans (n = 582) during a 24-week, randomized, open-label trial of methadone or buprenorphine/naloxone (Suboxone) for the treatment of opioid dependence. A single haplotype, tagged by rs10485058, was significantly associated with patient urinalysis data in the methadone treatment group. Methadone patients with the A/A genotype at rs10485058 were less likely to have opioid-positive urine drug screens than those in the combined A/G and G/G genotypes group (Relative Risk = 0.76, 95% confidence intervals = 0.73–0.80, p = 0.0064). Genotype at rs10485058 also predicted self-reported relapse rates in an independent population of Australian patients of European descent (n = 1215) who were receiving opioid substitution therapy (p = 0.003). In silico analysis predicted that miR-95-3p would interact with the G, but not the A allele of rs10485058. Luciferase assays indicated miR-95-3p decreased reporter activity of constructs containing the G, but not the A allele of rs10485058, suggesting a potential mechanism for the observed pharmacogenetic effect. These findings suggest that selection of a medication for opioid dependence based on rs10485058 genotype might improve outcomes in this ethnic group.
Collapse
Affiliation(s)
- R C Crist
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania School of Medicine, PA, Pennsylvania, USA
| | - G A Doyle
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania School of Medicine, PA, Pennsylvania, USA
| | - E C Nelson
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - L Degenhardt
- National Drug and Alcohol Research Centre, UNSW Australia, Sydney, New South Wales, Australia
| | - N G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - G W Montgomery
- The University of Queensland, Herston, Queensland, Australia
| | - A J Saxon
- Veteran's Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - W Ling
- University of California, Los Angeles, Integrated Substance Abuse Programs, Los Angeles, CA, USA
| | - W H Berrettini
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania School of Medicine, PA, Pennsylvania, USA
| |
Collapse
|
28
|
Kalsi G, Euesden J, Coleman JRI, Ducci F, Aliev F, Newhouse SJ, Liu X, Ma X, Wang Y, Collier DA, Asherson P, Li T, Breen G. Genome-Wide Association of Heroin Dependence in Han Chinese. PLoS One 2016; 11:e0167388. [PMID: 27936112 PMCID: PMC5147879 DOI: 10.1371/journal.pone.0167388] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 11/14/2016] [Indexed: 02/05/2023] Open
Abstract
Drug addiction is a costly and recurring healthcare problem, necessitating a need to understand risk factors and mechanisms of addiction, and to identify new biomarkers. To date, genome-wide association studies (GWAS) for heroin addiction have been limited; moreover they have been restricted to examining samples of European and African-American origin due to difficulty of recruiting samples from other populations. This is the first study to test a Han Chinese population; we performed a GWAS on a homogeneous sample of 370 Han Chinese subjects diagnosed with heroin dependence using the DSM-IV criteria and 134 ethnically matched controls. Analysis using the diagnostic criteria of heroin dependence yielded suggestive evidence for association between variants in the genes CCDC42 (coiled coil domain 42; p = 2.8x10-7) and BRSK2 (BR serine/threonine 2; p = 4.110−6). In addition, we found evidence for risk variants within the ARHGEF10 (Rho guanine nucleotide exchange factor 10) gene on chromosome 8 and variants in a region on chromosome 20q13, which is gene-poor but has a concentration of mRNAs and predicted miRNAs. Gene-based association analysis identified genome-wide significant association between variants in CCDC42 and heroin addiction. Additionally, when we investigated shared risk variants between heroin addiction and risk of other addiction-related and psychiatric phenotypes using polygenic risk scores, we found a suggestive relationship with variants predicting tobacco addiction, and a significant relationship with variants predicting schizophrenia. Our genome wide association study of heroin dependence provides data in a novel sample, with functionally plausible results and evidence of genetic data of value to the field.
Collapse
Affiliation(s)
- Gursharan Kalsi
- Institute of Psychiatry, Psychology and Neuroscience, MRC SGDP Centre, King’s College London, United Kingdom
| | - Jack Euesden
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Jonathan R. I. Coleman
- Institute of Psychiatry, Psychology and Neuroscience, MRC SGDP Centre, King’s College London, United Kingdom
| | - Francesca Ducci
- Institute of Psychiatry, Psychology and Neuroscience, MRC SGDP Centre, King’s College London, United Kingdom
| | - Fazil Aliev
- Department of Actuarial Sciences and Risk Management, Faculty of Business, Karabuk University, Karabuk, Turkey
| | - Stephen J. Newhouse
- Institute of Psychiatry, Psychology and Neuroscience, MRC SGDP Centre, King’s College London, United Kingdom
| | - Xiehe Liu
- Mental Health Center, West China Hospital, Sichuan University, Sichuan, People’s Republic of China
- Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, People’s Republic of China
| | - Xiaohong Ma
- Mental Health Center, West China Hospital, Sichuan University, Sichuan, People’s Republic of China
- Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, People’s Republic of China
| | - Yingcheng Wang
- Mental Health Center, West China Hospital, Sichuan University, Sichuan, People’s Republic of China
- Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, People’s Republic of China
| | - David A. Collier
- Institute of Psychiatry, Psychology and Neuroscience, MRC SGDP Centre, King’s College London, United Kingdom
- Lilly UK, Erl Wood Manor, Windlesham, Surrey, United Kingdom
| | - Philip Asherson
- Institute of Psychiatry, Psychology and Neuroscience, MRC SGDP Centre, King’s College London, United Kingdom
| | - Tao Li
- Department of Psychiatry, West China Hospital, School of Medicine, Sichuan University, Sichuan, People’s Republic of China
| | - Gerome Breen
- Institute of Psychiatry, Psychology and Neuroscience, MRC SGDP Centre, King’s College London, United Kingdom
- * E-mail:
| |
Collapse
|
29
|
Duquette LL, Mattiace F, Blum K, Waite RL, Boland T, McLaughlin T, Dushaj K, Febo M, Badgaiyan RD. Neurobiology of KB220Z-Glutaminergic-Dopaminergic Optimization Complex [GDOC] as a Liquid Nano: Clinical Activation of Brain in a Highly Functional Clinician Improving Focus, Motivation and Overall Sensory Input Following Chronic Intake. ACTA ACUST UNITED AC 2016; 3. [PMID: 29214221 PMCID: PMC5714519 DOI: 10.23937/2378-3656/1410104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background With neurogenetic and epigenetic tools utilized in research and neuroimaging, we are unraveling the mysteries of brain function, especially as it relates to Reward Deficiency (RDS). We encourage the development of pharmaceuticals or nutraceuticals that promote a reduction in dopamine resistance and balance brain neurochemistry, leading to dopamine homeostasis. We disclose self-assessment of a highly functional professional under work-related stress following KB220Z use, a liquid (aqua) nano glutaminergic-dopaminergic optimization complex (GDOC). Case presentation Subject took GDOC for one month. Subject self-administered GDOC using one-half-ounce twice a day. During first three days, unique brain activation occurred; resembling white noise after 30 minutes and sensation was strong for 45 minutes and then dissipated. He described effect as if his eyesight improved slightly and pointed out that his sense of smell and sleep greatly improved. Subject experienced a calming effect similar to meditation that could be linked to dopamine release. He also reported control of going over the edge after a hard day’s work, which was coupled with a slight increase in energy, increased motivation to work, increased focus and multi-tasking, with clearer purpose of task at hand. Subject felt less inhibited in a social setting and suggested Syndrome that GDOC increased his Behavior Activating System (reward), while having a decrease in the Behavior Inhibition System (caution). Conclusion These results and other related studies reveal an improved mood, work-related focus, and sleep. These effects as a subjective feeling of brain activation maybe due to direct or indirect dopaminergic interaction. While this case is encouraging, we must await more research in a larger randomized placebo-controlled study to map the role of GDOC, especially in a nano-sized product, to determine the possible effects on circuit inhibitory control and memory banks and the induction of dopamine homeostasis independent of either hypo- or hyper-dopaminergic traits/states.
Collapse
Affiliation(s)
- Lucien L Duquette
- New Pathway Counseling Services Inc., Paramus, NJ, USA.,Behavior Wellness Center, Englewood, NJ, USA
| | | | - Kenneth Blum
- Department of Psychiatry & McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, USA.,Division of Addiction Services, Dominion Diagnostics, LLC., North Kingstown, RI, USA.,Division of Neuroscience-Based Therapy, Summit Estate Recovery Center, Los Gatos, CA, USA.,Department of Psychiatry & Behavioral Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA.,Department of Clinical Neurology, PATH Foundation NY, New York, NY, USA.,Department of Nutrigenomic Translational Research, LaVita RDS, Salt Lake City, UT, USA.,Division of Neuroscience Research & Addiction Therapy, Shores Treatment & Recovery Center, Port Saint Lucie, FL, USA
| | - Roger L Waite
- Department of Nutrigenomic Translational Research, LaVita RDS, Salt Lake City, UT, USA
| | | | | | - Kristina Dushaj
- Department of Clinical Neurology, PATH Foundation NY, New York, NY, USA
| | - Marcelo Febo
- Department of Psychiatry & McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, USA
| | - Rajendra D Badgaiyan
- Department of Psychiatry, Laboratory of Molecular and Functional Imaging, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
30
|
Bandelow B, Wedekind D. Possible role of a dysregulation of the endogenous opioid system in antisocial personality disorder. Hum Psychopharmacol 2015; 30:393-415. [PMID: 26250442 DOI: 10.1002/hup.2497] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/29/2015] [Accepted: 06/23/2015] [Indexed: 11/09/2022]
Abstract
Around half the inmates in prison institutions have antisocial personality disorder (ASPD). A recent theory has proposed that a dysfunction of the endogenous opioid system (EOS) underlies the neurobiology of borderline personality disorder (BPD). In the present theoretical paper, based on a comprehensive database and hand search of the relevant literature, this hypothesis is extended to ASPD, which may be the predominant expression of EOS dysfunction in men, while the same pathology underlies BPD in women. According to evidence from human and animal studies, the problematic behaviours of persons with antisocial, callous, or psychopathic traits may be seen as desperate, unconscious attempts to stimulate their deficient EOS, which plays a key role in brain reward circuits. If the needs of this system are not being met, the affected persons experience dysphoric mood, discomfort, or irritability, and strive to increase binding of endogenous opioids to receptors by using the rewarding effects of aggression by exertion of physical or manipulative power on others, by abusing alcohol or substances that have the reward system as target, by creating an "endorphin rush" by self-harm, by increasing the frequency of their sexual contacts, or by impulsive actions and sensation seeking. Symptoms associated with ASPD can be treated with opioid antagonists like naltrexone, naloxone, or nalmefene.
Collapse
Affiliation(s)
- Borwin Bandelow
- Department of Psychiatry and Psychotherapy, University of Göttingen, Germany
| | - Dirk Wedekind
- Department of Psychiatry and Psychotherapy, University of Göttingen, Germany
| |
Collapse
|
31
|
Jian CD, Huang JM, Meng LQ, Li XB, Huang RY, Shi SL, Wu Y, Qin C, Chen J, Zhang YM, Wang S, Feng YL, Zhou SN. SNCA rs3822086 C>T Polymorphism Increases the Susceptibility to Parkinson's Disease in a Chinese Han Population. Genet Test Mol Biomarkers 2015. [PMID: 26203864 DOI: 10.1089/gtmb.2015.0046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Chong-Dong Jian
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jian-Min Huang
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Lan-Qing Meng
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xue-Bin Li
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Rui-Ya Huang
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Sheng-Liang Shi
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yuan Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chao Qin
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jin Chen
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yan-Mei Zhang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shuang Wang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yin-Ling Feng
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Sheng-Nian Zhou
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
32
|
Salvatore JE, Edwards AC, McClintick JN, Bigdeli TB, Adkins A, Aliev F, Edenberg HJ, Foroud T, Hesselbrock V, Kramer J, Nurnberger JI, Schuckit M, Tischfield JA, Xuei X, Dick DM. Genome-wide association data suggest ABCB1 and immune-related gene sets may be involved in adult antisocial behavior. Transl Psychiatry 2015; 5:e558. [PMID: 25918995 PMCID: PMC4462601 DOI: 10.1038/tp.2015.36] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/09/2015] [Indexed: 11/09/2022] Open
Abstract
Adult antisocial behavior (AAB) is moderately heritable, relatively common and has adverse consequences for individuals and society. We examined the molecular genetic basis of AAB in 1379 participants from a case-control study in which the cases met criteria for alcohol dependence. We also examined whether genes of interest were expressed in human brain. AAB was measured using a count of the number of Antisocial Personality Disorder criteria endorsed under criterion A from the Diagnostic and Statistical Manual of Mental Disorders, 4th Edition (DSM-IV). Participants were genotyped on the Illumina Human 1M BeadChip. In total, all single-nucleotide polymorphisms (SNPs) accounted for 25% of the variance in AAB, although this estimate was not significant (P=0.09). Enrichment tests indicated that more significantly associated genes were over-represented in seven gene sets, and most were immune related. Our most highly associated SNP (rs4728702, P=5.77 × 10(-7)) was located in the protein-coding adenosine triphosphate-binding cassette, sub-family B (MDR/TAP), member 1 (ABCB1). In a gene-based test, ABCB1 was genome-wide significant (q=0.03). Expression analyses indicated that ABCB1 was robustly expressed in the brain. ABCB1 has been implicated in substance use, and in post hoc tests we found that variation in ABCB1 was associated with DSM-IV alcohol and cocaine dependence criterion counts. These results suggest that ABCB1 may confer risk across externalizing behaviors, and are consistent with previous suggestions that immune pathways are associated with externalizing behaviors. The results should be tempered by the fact that we did not replicate the associations for ABCB1 or the gene sets in a less-affected independent sample.
Collapse
Affiliation(s)
- J E Salvatore
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - A C Edwards
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - J N McClintick
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
| | - T B Bigdeli
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - A Adkins
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - F Aliev
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
- Department of Statistics and Institute of Biotechnology, Ankara University, Ankara, Turkey
| | - H J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
| | - T Foroud
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| | - V Hesselbrock
- Department of Psychiatry, University of Connecticut, Farmington, CT, USA
| | - J Kramer
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - J I Nurnberger
- Department of Psychiatry, Indiana University, Indianapolis, IN, USA
| | - M Schuckit
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - J A Tischfield
- Department of Genetics, Rutgers University, Piscataway, NJ, USA
| | - X Xuei
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
| | - D M Dick
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
33
|
Ahn WY, Vasilev G, Lee SH, Busemeyer JR, Kruschke JK, Bechara A, Vassileva J. Decision-making in stimulant and opiate addicts in protracted abstinence: evidence from computational modeling with pure users. Front Psychol 2014; 5:849. [PMID: 25161631 PMCID: PMC4129374 DOI: 10.3389/fpsyg.2014.00849] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/17/2014] [Indexed: 12/04/2022] Open
Abstract
Substance dependent individuals (SDI) often exhibit decision-making deficits; however, it remains unclear whether the nature of the underlying decision-making processes is the same in users of different classes of drugs and whether these deficits persist after discontinuation of drug use. We used computational modeling to address these questions in a unique sample of relatively “pure” amphetamine-dependent (N = 38) and heroin-dependent individuals (N = 43) who were currently in protracted abstinence, and in 48 healthy controls (HC). A Bayesian model comparison technique, a simulation method, and parameter recovery tests were used to compare three cognitive models: (1) Prospect Valence Learning with decay reinforcement learning rule (PVL-DecayRI), (2) PVL with delta learning rule (PVL-Delta), and (3) Value-Plus-Perseverance (VPP) model based on Win-Stay-Lose-Switch (WSLS) strategy. The model comparison results indicated that the VPP model, a hybrid model of reinforcement learning (RL) and a heuristic strategy of perseverance had the best post-hoc model fit, but the two PVL models showed better simulation and parameter recovery performance. Computational modeling results suggested that overall all three groups relied more on RL than on a WSLS strategy. Heroin users displayed reduced loss aversion relative to HC across all three models, which suggests that their decision-making deficits are longstanding (or pre-existing) and may be driven by reduced sensitivity to loss. In contrast, amphetamine users showed comparable cognitive functions to HC with the VPP model, whereas the second best-fitting model with relatively good simulation performance (PVL-DecayRI) revealed increased reward sensitivity relative to HC. These results suggest that some decision-making deficits persist in protracted abstinence and may be mediated by different mechanisms in opiate and stimulant users.
Collapse
Affiliation(s)
- Woo-Young Ahn
- Virginia Tech Carilion Research Institute, Virginia Tech Roanoke, VA, USA
| | | | - Sung-Ha Lee
- Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, USA
| | - Jerome R Busemeyer
- Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, USA
| | - John K Kruschke
- Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, USA
| | - Antoine Bechara
- Department of Psychology, University of Southern California Los Angeles, CA, USA ; Brain and Creativity Institute, University of Southern California Los Angeles, CA, USA
| | - Jasmin Vassileva
- Department of Psychiatry, Virginia Commonwealth University School of Medicine Richmond, VA, USA
| |
Collapse
|
34
|
Epigenetically modified nucleotides in chronic heroin and cocaine treated mice. Toxicol Lett 2014; 229:451-7. [PMID: 25064621 DOI: 10.1016/j.toxlet.2014.07.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/21/2014] [Accepted: 07/22/2014] [Indexed: 12/19/2022]
Abstract
Epigenetic changes include the addition of a methyl group to the 5' carbon of the cytosine ring, known as DNA methylation, which results in the generation of the fifth DNA base, namely 5-methylcytosine. During active or passive demethylation, an intermediate modified base is formed, 5-hydroxymethylcytosine. We have currently quantified 5-methylcytosine and 5-hydroxymethylcytosine in the liver and brain of mice treated with cocaine or heroin, using liquid chromatography/tandem mass spectrometry (LC-MS/MS). Our results show that global 5-methylcytosine levels are not affected by heroin or cocaine administration, neither in the liver nor in the brain. However, 5-hydroxymethylcytosine levels are reduced in the liver following cocaine administration, while they are not affected by cocaine in the brain or by heroin administration in the liver and the brain. Elucidation of the epigenetic phenomena that takes place with respect to drug abuse and addiction, via quantitative analysis of different modified bases, may enable a better understanding of the underlying mechanisms and may lead to more personalized and effective treatment options.
Collapse
|
35
|
Enabah D, El Baz H, Moselhy H. Higher frequency of C.3435 of the ABCB1 gene in patients with tramadol dependence disorder. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2014; 40:317-20. [PMID: 24950410 DOI: 10.3109/00952990.2014.925468] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Polymorphic variation at the ABCB1 gene has been shown to affect the pharmacodynamics and kinetics of various drugs. AIM This study aimed to determine the frequency of occurrence of Single Nucleotide Polymorphism (SNP) in position A118G OPRM1 (rs1799971) gene and C.3435 (rs1045642) gene in tramadol users in comparison with normal controls. METHODS This was a cross sectional case-control outpatient study. The study sample consisted of 127 subjects (74 tramadol-dependents and 50 healthy controls). All patients fulfilled the Diagnostic and Statistical Manual IV Criteria for substance dependence (on tramadol). Genotyping of the OPRM1 gene 118 SNP and ABCB1 genes C.3435 SNP was performed by PCR, followed by restriction fragment length polymorphism identification. RESULTS A significant association was found between the ABCB1 gene T allele at the polymorphic site 3435 and tramadol dependence. No significant association was observed with the A118G OPRM1 gene. CONCLUSION The high frequency of ABCB1 gene T allele present at the polymorphic site 3435 could provide a protective mechanism from tramadol dependence disorder. Further study, using a larger sample, would be useful in further evaluating the possible role of ABCB1 gene polymorphisms.
Collapse
|
36
|
Mistry CJ, Bawor M, Desai D, Marsh DC, Samaan Z. Genetics of Opioid Dependence: A Review of the Genetic Contribution to Opioid Dependence. CURRENT PSYCHIATRY REVIEWS 2014; 10:156-167. [PMID: 25242908 PMCID: PMC4155832 DOI: 10.2174/1573400510666140320000928] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 08/14/2013] [Accepted: 01/15/2014] [Indexed: 02/08/2023]
Abstract
This narrative review aims to provide an overview of the impact of opioid dependence and the contribution of genetics to opioid dependence. Epidemiological data demonstrate that opioid dependence is a global trend with far-reaching effects on the social, economic, and health care systems. A review of classical genetic studies of opioid use suggests significant heritability of drug use behavior, however the evidence from molecular genetic studies is inconclusive. Nonetheless, certain genetic variants are important to consider given their role in the pathophysiology of addictive behavior. We undertook a literature review to identify the current state of knowledge regarding the role of genes in opioid dependence. Determining the association of genetic markers could change the current understanding of the various factors contributing to opioid dependence and therefore may improve recognition of individuals at risk for the disorder and prevention and treatment strategies.
Collapse
Affiliation(s)
- Chetna J Mistry
- Arts & Science Undergraduate Program, McMaster University, ON, Canada
| | - Monica Bawor
- McMaster Integrative Neuroscience Discovery & Study (MiNDS), McMaster University, ON, Canada
| | - Dipika Desai
- Population Health Research Institute, Hamilton, ON, Canada
| | - David C Marsh
- Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Zainab Samaan
- Population Health Research Institute, Hamilton, ON, Canada
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, ON, Canada
- Population Genomics Program, Chanchlani Research Centre, McMaster University, ON, Canada
- Department of Clinical Epidemiology and Biostatistics, McMaster University, ON, Canada
| |
Collapse
|
37
|
Haerian BS, Haerian MS. OPRM1 rs1799971 polymorphism and opioid dependence: evidence from a meta-analysis. Pharmacogenomics 2014; 14:813-24. [PMID: 23651028 DOI: 10.2217/pgs.13.57] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The OPRM1 gene encodes the µ-opioid receptor, which is the primary site of action of most opioids. Several studies and three meta-analyses have examined a possible link between the exonic OPRM1 A118G (rs1799971) polymorphism and opioid dependence; however, results have been inconclusive. Therefore, a systematic review and meta-analysis have been carried out to examine whether this polymorphism is associated with opioid dependence. Thirteen studies (n = 9385), comprising 4601 opioid dependents and 4784 controls, which evaluated association of the OPRM1 rs1799971 polymorphism with susceptibility to opioids, were included in this study. Our meta-analysis showed significant association between this polymorphism and susceptibility to opioid dependence in overall studies under a codominant model, as well as susceptibility to opioid dependence or heroin dependence in Asians under an autosomal dominant model. The nonsynonymous OPRM1 rs1799971 might be a risk factor for addiction to opioids or heroin in an Asian population.
Collapse
Affiliation(s)
- Batoul Sadat Haerian
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | | |
Collapse
|
38
|
Bough KJ, Amur S, Lao G, Hemby SE, Tannu NS, Kampman KM, Schmitz JM, Martinez D, Merchant KM, Green C, Sharma J, Dougherty AH, Moeller FG. Biomarkers for the development of new medications for cocaine dependence. Neuropsychopharmacology 2014; 39:202-19. [PMID: 23979119 PMCID: PMC3857653 DOI: 10.1038/npp.2013.210] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 06/19/2013] [Accepted: 07/01/2013] [Indexed: 01/20/2023]
Abstract
There has been significant progress in personalized drug development. In large part, this has taken place in the oncology field and been due to the ability of researchers/clinicians to discover and develop novel drug development tools (DDTs), such as biomarkers. In cancer treatment research, biomarkers have permitted a more accurate pathophysiological characterization of an individual patient, and have enabled practitioners to target mechanistically the right drug, to the right patient, at the right time. Similar to cancer, patients with substance use disorders (SUDs) present clinically with heterogeneous symptomatology and respond variably to therapeutic interventions. If comparable biomarkers could be identified and developed for SUDs, significant diagnostic and therapeutic advances could be made. In this review, we highlight current opportunities and difficulties pertaining to the identification and development of biomarkers for SUDs. We focus on cocaine dependence as an example. Putative diagnostic, pharmacodynamic (PD), and predictive biomarkers for cocaine dependence are discussed across a range of methodological approaches. A possible cocaine-dependent clinical outcome assessment (COA)--another type of defined DDT--is also discussed. At present, biomarkers for cocaine dependence are in their infancy. Much additional research will be needed to identify, validate, and qualify these putative tools prior to their potential use for medications development and/or application to clinical practice. However, with a large unmet medical need and an estimated market size of several hundred million dollars per year, if developed, biomarkers for cocaine dependence will hold tremendous value to both industry and public health.
Collapse
Affiliation(s)
- Kristopher J Bough
- Division of Basic Neuroscience and Behavioral Research, National Institute on Drug Abuse, Bethesda, MD, USA
| | - Shashi Amur
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Guifang Lao
- Division of Pharmacotherapies and Medical Consequences, National Institute on Drug Abuse, Bethesda, MD, USA
| | - Scott E Hemby
- Department of Physiology & Pharmacology, Wake Forest University, Winston-Salem, NC, USA
| | - Nilesh S Tannu
- Department of Psychiatry and Behavioral Sciences, University of Texas—Houston Medical School, Houston, TX, USA
| | - Kyle M Kampman
- Department of Psychiatry, University of Pennsylvania—School of Medicine, Philadelphia, PA, USA
| | - Joy M Schmitz
- Department of Psychiatry and Behavioral Sciences, University of Texas—Houston Medical School, Houston, TX, USA
| | - Diana Martinez
- Department of Psychiatry, Columbia University/New York State University, New York, NY, USA
| | | | - Charles Green
- Department of Pediatrics, University of Texas—Houston Medical School, Houston, TX, USA
| | - Jyoti Sharma
- Department of Cardiovascular Medicine, University of Texas—Houston Medical School, Houston, TX, USA
| | - Anne H Dougherty
- Department of Cardiovascular Medicine, University of Texas—Houston Medical School, Houston, TX, USA
| | - F Gerard Moeller
- Department of Psychiatry and Pharmacology and Toxicology, Virginia Commonwealth University Medical School, Richmond, VA, USA
| |
Collapse
|
39
|
Hauser KF, Knapp PE. Interactions of HIV and drugs of abuse: the importance of glia, neural progenitors, and host genetic factors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 118:231-313. [PMID: 25175867 PMCID: PMC4304845 DOI: 10.1016/b978-0-12-801284-0.00009-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Considerable insight has been gained into the comorbid, interactive effects of HIV and drug abuse in the brain using experimental models. This review, which considers opiates, methamphetamine, and cocaine, emphasizes the importance of host genetics and glial plasticity in driving the pathogenic neuron remodeling underlying neuro-acquired immunodeficiency syndrome and drug abuse comorbidity. Clinical findings are less concordant than experimental work, and the response of individuals to HIV and to drug abuse can vary tremendously. Host-genetic variability is important in determining viral tropism, neuropathogenesis, drug responses, and addictive behavior. However, genetic differences alone cannot account for individual variability in the brain "connectome." Environment and experience are critical determinants in the evolution of synaptic circuitry throughout life. Neurons and glia both exercise control over determinants of synaptic plasticity that are disrupted by HIV and drug abuse. Perivascular macrophages, microglia, and to a lesser extent astroglia can harbor the infection. Uninfected bystanders, especially astroglia, propagate and amplify inflammatory signals. Drug abuse by itself derails neuronal and glial function, and the outcome of chronic exposure is maladaptive plasticity. The negative consequences of coexposure to HIV and drug abuse are determined by numerous factors including genetics, sex, age, and multidrug exposure. Glia and some neurons are generated throughout life, and their progenitors appear to be targets of HIV and opiates/psychostimulants. The chronic nature of HIV and drug abuse appears to result in sustained alterations in the maturation and fate of neural progenitors, which may affect the balance of glial populations within multiple brain regions.
Collapse
Affiliation(s)
- Kurt F Hauser
- Department of Pharmacology & Toxicology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA.
| | - Pamela E Knapp
- Department of Pharmacology & Toxicology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA; Department of Anatomy & Neurobiology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
40
|
Beer B, Erb R, Pavlic M, Ulmer H, Giacomuzzi S, Riemer Y, Oberacher H. Association of polymorphisms in pharmacogenetic candidate genes (OPRD1, GAL, ABCB1, OPRM1) with opioid dependence in European population: a case-control study. PLoS One 2013; 8:e75359. [PMID: 24086514 PMCID: PMC3783401 DOI: 10.1371/journal.pone.0075359] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/13/2013] [Indexed: 11/18/2022] Open
Abstract
It is becoming increasingly evident that genetic variants contribute to the development of opioid addiction. An elucidation of these genetic factors is crucial for a better understanding of this chronic disease and may help to develop novel therapeutic strategies. In recent years, several candidate genes were implicated in opioid dependence. However, most study findings have not been replicated and additional studies are required before reported associations can be considered robust. Thus, the major objective of this study was to replicate earlier findings and to identify new genetic polymorphisms contributing to the individual susceptibility to opioid addiction, respectively. Therefore, a candidate gene association study was conducted including 142 well-phenotyped long-term opioid addicts undergoing opioid maintenance therapy and 142 well-matched healthy controls. In both study groups, 24 single nucleotide polymorphisms predominantly located in pharmacogenetic candidate genes have been genotyped using an accurate mass spectrometry based method. The most significant associations with opioid addiction (remaining significant after adjustment for multiple testing) were observed for the rs948854 SNP in the galanin gene (GAL, p = 0.001) and the rs2236861 SNP in the delta opioid receptor gene (OPRD1, p = 0.001). Moreover, an association of the ATP binding cassette transporter 1 (ABCB1) variant rs1045642 and the Mu Opioid receptor (OPRM1) variant rs9479757 with opioid addiction was observed. The present study provides further support for a contribution of GAL and OPRD1 variants to the development of opioid addiction. Furthermore, our results indicate a potential contribution of OPRM1 and ABCB1 SNPs to the development of this chronic relapsing disease. Therefore it seems important that these genes are addressed in further addiction related studies.
Collapse
Affiliation(s)
- Beate Beer
- Institute of Legal Medicine, Innsbruck Medical University, Innsbruck, Austria
| | - Robert Erb
- Institute of Legal Medicine, Innsbruck Medical University, Innsbruck, Austria
| | - Marion Pavlic
- Institute of Legal Medicine, Innsbruck Medical University, Innsbruck, Austria
| | - Hanno Ulmer
- Department of Medical Statistics, Informatics, and Health Economics, Innsbruck Medical University, Innsbruck, Austria
| | - Salvatore Giacomuzzi
- Department of General Psychiatry, Innsbruck Medical University, Innsbruck, Austria
| | - Yvonne Riemer
- Department of General Psychiatry, Innsbruck Medical University, Innsbruck, Austria
| | - Herbert Oberacher
- Institute of Legal Medicine, Innsbruck Medical University, Innsbruck, Austria
- * E-mail:
| |
Collapse
|
41
|
Greenwald MK, Steinmiller CL, Sliwerska E, Lundahl L, Burmeister M. BDNF Val(66)Met genotype is associated with drug-seeking phenotypes in heroin-dependent individuals: a pilot study. Addict Biol 2013; 18:836-45. [PMID: 22339949 DOI: 10.1111/j.1369-1600.2011.00431.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) Val(66)Met genotype has been associated with neurobehavioral deficits. To examine its relevance for addiction, we examined BDNF genotype differences in drug-seeking behavior. Heroin-dependent volunteers (n = 128) completed an interview that assessed past-month naturalistic drug-seeking/use behaviors. In African Americans (n = 74), the Met allele was uncommon (carrier frequency 6.8%); thus, analyses focused on European Americans (n = 54), in whom the Met allele was common (carrier frequency 37.0%). In their natural setting, Met carriers (n = 20) reported more time- and cost-intensive heroin-seeking and more cigarette use than Val homozygotes (n = 34). BDNF Val(66)Met genotype predicted 18.4% of variance in 'weekly heroin investment' (purchasing time × amount × frequency). These data suggest that the BDNF Met allele may confer a 'preferred drug-invested' phenotype, resistant to moderating effects of higher drug prices and non-drug reinforcement. These preliminary hypothesis-generating findings require replication, but are consistent with pre-clinical data that demonstrate neurotrophic influence in drug reinforcement. Whether this genotype is relevant to other abused substances besides opioids or nicotine, or treatment response, remains to be determined.
Collapse
Affiliation(s)
- Mark K Greenwald
- Substance Abuse Research Division, Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA.
| | | | | | | | | |
Collapse
|
42
|
Bruehl S, Apkarian AV, Ballantyne JC, Berger A, Borsook D, Chen WG, Farrar JT, Haythornthwaite JA, Horn SD, Iadarola MJ, Inturrisi CE, Lao L, Mackey S, Mao J, Sawczuk A, Uhl GR, Witter J, Woolf CJ, Zubieta JK, Lin Y. Personalized medicine and opioid analgesic prescribing for chronic pain: opportunities and challenges. THE JOURNAL OF PAIN 2013; 14:103-13. [PMID: 23374939 DOI: 10.1016/j.jpain.2012.10.016] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/17/2012] [Accepted: 10/25/2012] [Indexed: 01/05/2023]
Abstract
UNLABELLED Use of opioid analgesics for pain management has increased dramatically over the past decade, with corresponding increases in negative sequelae including overdose and death. There is currently no well-validated objective means of accurately identifying patients likely to experience good analgesia with low side effects and abuse risk prior to initiating opioid therapy. This paper discusses the concept of data-based personalized prescribing of opioid analgesics as a means to achieve this goal. Strengths, weaknesses, and potential synergism of traditional randomized placebo-controlled trial (RCT) and practice-based evidence (PBE) methodologies as means to acquire the clinical data necessary to develop validated personalized analgesic-prescribing algorithms are overviewed. Several predictive factors that might be incorporated into such algorithms are briefly discussed, including genetic factors, differences in brain structure and function, differences in neurotransmitter pathways, and patient phenotypic variables such as negative affect, sex, and pain sensitivity. Currently available research is insufficient to inform development of quantitative analgesic-prescribing algorithms. However, responder subtype analyses made practical by the large numbers of chronic pain patients in proposed collaborative PBE pain registries, in conjunction with follow-up validation RCTs, may eventually permit development of clinically useful analgesic-prescribing algorithms. PERSPECTIVE Current research is insufficient to base opioid analgesic prescribing on patient characteristics. Collaborative PBE studies in large, diverse pain patient samples in conjunction with follow-up RCTs may permit development of quantitative analgesic-prescribing algorithms that could optimize opioid analgesic effectiveness and mitigate risks of opioid-related abuse and mortality.
Collapse
Affiliation(s)
- Stephen Bruehl
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
BACKGROUND The clinical utility of opioids is limited by adverse drug effects including respiratory depression, sedation, nausea, and pruritus. In addition, abuse of prescription opioids is problematic. Gaining a better understanding of the genetic and environmental mechanisms contributing to an individual's susceptibility to adverse opioid effects is essential to identify patients at risk. METHODS A classic twin study paradigm provided estimates for the genetic and familial (genetic and/or shared environment) contribution to acute adverse and affective opioid responses, all secondary outcomes of a larger dataset. One hundred twenty-one twin pairs were recruited in a single occasion, randomized, double-blind, and placebo-controlled study. The μ-opioid receptor agonist alfentanil and saline placebo were administered as target-controlled infusions under carefully monitored laboratory conditions. Measured outcomes included respiratory depression, sedation, nausea, pruritus, drug liking, and drug disliking. Demographic information was collected, and aspects of mood and sleep were evaluated. RESULTS Significant heritability was detected for respiratory depression (30%), nausea (59%), and drug disliking (36%). Significant familial effects were detected for sedation (29%), pruritus (38%), dizziness (32%), and drug liking (26%). Significant covariates included age, sex, race, ethnicity, education, mood, and depression. Covariates affected sedation, pruritus, drug liking and disliking, and dizziness. CONCLUSIONS This study demonstrates that large-scale efforts to collect quantitative and well-defined opioid response data are not only feasible but also produce data that are suitable for genetic analysis. Genetic, environmental, and demographic factors work together to control adverse and reinforcing opioid responses, but contribute differently to specific responses.
Collapse
|
44
|
Wang TY, Lee SY, Chen SL, Chang YH, Chen SH, Chu CH, Huang SY, Tzeng NS, Wang CL, Yeh PH, Lee IH, Yeh TL, Yang YK, Lu RB. The ADH1B and DRD2 gene polymorphism may modify the protective effect of the ALDH2 gene against heroin dependence. Prog Neuropsychopharmacol Biol Psychiatry 2013; 43:134-9. [PMID: 23266708 DOI: 10.1016/j.pnpbp.2012.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/10/2012] [Accepted: 12/15/2012] [Indexed: 10/27/2022]
Abstract
Understanding the influences of genes involved in dopamine and serotonin metabolism, such as the aldehyde dehydrogenase 2 (ALDH2) and alcohol dehydrogenase 1B (ADH1B) genes, is critical for understanding addictive behavior. In addition, dopamine D2 receptor (DRD2) gene may also interact with the dopamine metabolizing genes and link to addiction. Therefore, we investigated the association between the ALDH2, ADH1B and DRD2 polymorphisms and heroin dependence. Heroin-dependent Han Chinese patients (n=304) and healthy controls (n=335) were recruited. Genotypes of ALDH2, ADH1B and DRD2 polymorphisms were analyzed using a polymerase chain reaction with restriction fragment length polymorphism. The frequency of the ALDH2*1/*1 genotype was significantly lower in heroin-dependent patients than in controls, but the frequency of ADH1B and DRD2 genotypes was not significantly different. Further stratification of the ALDH2 gene with the ADH1B gene showed that the protective effect of ALDH2*1/*1 existed only in patients who also carried the ADH1B*1/*1 and ADH1B*1/*2 genotype. Logistic regression analysis showed a significant interaction between ALDH2 and ADH1B (P=0.022) and DRD2, ALDH2 and ADH1B in patients (P=0.037). The ALDH2*1/*1, ADH1B*1/*1, and ADH1B*1/*2 genotypes may interact and protect their carriers against heroin dependence and the protective effect may be varied by the DRD2 gene polymorphism. We conclude that the protective effect of the ALDH2 polymorphism against heroin dependence may be modified by the ADH1B and DRD2 polymorphism.
Collapse
Affiliation(s)
- Tzu-Yun Wang
- Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Carpentier PJ, Arias Vasquez A, Hoogman M, Onnink M, Kan CC, Kooij JJS, Makkinje R, Iskandar S, Kiemeney LA, de Jong CAJ, Franke B, Buitelaar JK. Shared and unique genetic contributions to attention deficit/hyperactivity disorder and substance use disorders: a pilot study of six candidate genes. Eur Neuropsychopharmacol 2013; 23:448-57. [PMID: 22841130 DOI: 10.1016/j.euroneuro.2012.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 06/01/2012] [Accepted: 07/09/2012] [Indexed: 11/16/2022]
Abstract
The shared genetic basis of attention deficit/hyperactivity disorder (ADHD) and substance use disorders (SUDs) was explored by investigating the association of candidate risk factors in neurotransmitter genes with both disorders. One hundred seven methadone maintenance treatment patients, 36 having an ADHD diagnosis, 176 adult patients with ADHD without SUDs, and 500 healthy controls were genotyped for variants in the DRD4 (exon 3 VNTR), DRD5 (upstream VNTR), HTR1B (rs6296), DBH (rs2519152), COMT (rs4680; Val158Met), and OPRM1 (rs1799971; 118A>G) genes. Association with disease was tested using logistic regression models. This pilot study was adequately powered to detect larger genetic effects (OR≥2) of risk alleles with a low frequency. Compared to controls, ADHD patients (with and without SUDs) showed significantly increased frequency of the DBH (rs2519152: OR 1.73; CI 1.15-2.59; P=0.008) and the OPRM1 risk genotypes (rs1799971: OR 1.71; CI 1.17-2.50; P=0.006). The DBH risk genotype was associated with ADHD diagnosis, with the association strongest in the pure ADHD group. The OPRM1 risk genotype increased the risk for the combined ADHD and SUD phenotype. The present study strengthens the evidence for a shared genetic basis for ADHD and addiction. The association of OPRM1 with the ADHD and SUD combination could help to explain the contradictory results of previous studies. The power limitations of the study restrict the significance of these findings: replication in larger samples is warranted.
Collapse
Affiliation(s)
- P J Carpentier
- Novadic-Kentron, Network for Addiction Treatment Services, Vught, The Netherlands; Reinier van Arkel groep, 's-Hertogenbosch, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Discovery of the inhibitory effect of a phosphatidylinositol derivative on P-glycoprotein by virtual screening followed by in vitro cellular studies. PLoS One 2013; 8:e60679. [PMID: 23593281 PMCID: PMC3621910 DOI: 10.1371/journal.pone.0060679] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/01/2013] [Indexed: 11/23/2022] Open
Abstract
P-glycoprotein is capable of effluxing a broad range of cytosolic and membrane penetrating xenobiotic substrates, thus leading to multi-drug resistance and posing a threat for the therapeutic treatment of several diseases, including cancer and central nervous disorders. Herein, a virtual screening campaign followed by experimental validation in Caco-2, MDKCII, and MDKCII mdr1 transfected cell lines has been conducted for the identification of novel phospholipids with P-gp transportation inhibitory activity. Phosphatidylinositol-(1,2-dioctanoyl)-sodium salt (8∶0 PI) was found to significantly inhibit transmembrane P-gp transportation in vitro in a reproducible-, cell line-, and substrate-independent manner. Further tests are needed to determine whether this and other phosphatidylinositols could be co-administered with oral drugs to successfully increase their bioavailability. Moreover, as phosphatidylinositols and phosphoinositides are present in the human diet and are known to play an important role in signal transduction and cell motility, our finding could be of substantial interest for nutrition science as well.
Collapse
|
47
|
Isaza C, Henao J, Beltrán L, Porras L, Gonzalez M, Cruz R, Carracedo A. Genetic variants associated with addictive behavior in Colombian addicted and non-addicted to heroin or cocaine. Colomb Med (Cali) 2013; 44:19-25. [PMID: 24892317 DOI: pmid/24892317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 09/09/2012] [Accepted: 02/11/2013] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Determine the prevalence and compare some genetic markers involved in addictive behavior in a group of addicts to derivative of coca (cocaine/crack) or heroin and a control group of non-addicted people matched for gender, age and ethnicity. METHODS A 120 addicts and 120 non-addicts Colombian male were surveyed and genotyped for 18 polymorphism of the OPRM1, DRD2, DRD4, SLC6A3, SLC6A4, ABCB1, DβH and CYP2B6 genes. For the identification of alleles markers were used mini-sequencing and fragment multiplex PCR techniques; ethnicity of cases and controls was analyzed with 61 AIMs. RESULTS The age of onset use of heroin or coca derivatives (cocaine/crack) was 16.5±6 years and 99.2% of them consume several illicit drugs. It showed that controls and addicts belong to the same ethnic group. Significant differences between addicts and controls in relation to schooling, marital status, social security family history of substance abuse (p <0.001), Int8-VNTR SLC6A3 gene (p= 0.015) and SNP 3435C>T ABCB1 gene (p= 0.001) were found. CONCLUSION The present results indicate that the VNTR- 6R polymorphism of the gene SLC6A3 and the genotype 3435CC in the ABCB1 gene, are both associated with addictive behavior to heroin or cocaine.
Collapse
Affiliation(s)
- Carlos Isaza
- Genetical Medical Laboratory, Pharmacogenetics Research Group, Universidad Tecnológica de Pereira, Facultad de Ciencias de la Salud, Pereira, Colombia, E-mails :
| | - Julieta Henao
- Genetical Medical Laboratory, Pharmacogenetics Research Group, Universidad Tecnológica de Pereira, Facultad de Ciencias de la Salud, Pereira, Colombia, E-mails :
| | - Leonardo Beltrán
- Genetical Medical Laboratory, Pharmacogenetics Research Group, Universidad Tecnológica de Pereira, Facultad de Ciencias de la Salud, Pereira, Colombia, E-mails :
| | - Liliana Porras
- Genetical Medical Laboratory, Pharmacogenetics Research Group, Universidad Tecnológica de Pereira, Facultad de Ciencias de la Salud, Pereira, Colombia, E-mails :
| | - Martha Gonzalez
- Genetical Medical Laboratory, Pharmacogenetics Research Group, Universidad Tecnológica de Pereira, Facultad de Ciencias de la Salud, Pereira, Colombia, E-mails :
| | - Raquel Cruz
- Institute of Forensic Science, Genomic Medicine Group-CIBERER, Universidad de Santiago de Compostela, Santiago de Compostela, Spain, E-mails:
| | - Angel Carracedo
- Institute of Forensic Science, Genomic Medicine Group-CIBERER, Universidad de Santiago de Compostela, Santiago de Compostela, Spain, E-mails:
| |
Collapse
|
48
|
Crist RC, Ambrose-Lanci LM, Vaswani M, Clarke TK, Zeng A, Yuan C, Ferraro TN, Hakonarson H, Kampman KM, Dackis CA, Pettinati HM, O'Brien CP, Oslin DW, Doyle GA, Lohoff FW, Berrettini WH. Case-control association analysis of polymorphisms in the δ-opioid receptor, OPRD1, with cocaine and opioid addicted populations. Drug Alcohol Depend 2013; 127:122-8. [PMID: 22795689 PMCID: PMC3509227 DOI: 10.1016/j.drugalcdep.2012.06.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 06/18/2012] [Accepted: 06/21/2012] [Indexed: 11/27/2022]
Abstract
BACKGROUND Addiction susceptibility and treatment responsiveness are greatly influenced by genetic factors. Sequence variation in genes involved in the mechanisms of drug action have the potential to influence addiction risk and treatment outcome. The opioid receptor system is involved in mediating the rewarding effects of cocaine and opioids. The μ-opioid receptor (MOR) has traditionally been considered the primary target for opioid addiction. The MOR, however, interacts with and is regulated by many known MOR interacting proteins (MORIPs), including the δ-opioid receptor (DOR). METHODS The present study evaluated the contribution of OPRD1, the gene encoding the DOR, to the risk of addiction to opioids and cocaine. The association of OPRD1 polymorphisms with both opioid addiction (OA) and cocaine addiction (CA) was analyzed in African American (OA n=336, CA n=503) and European American (OA n=1007, CA n=336) populations. RESULTS The primary finding of this study is an association of rs678849 with cocaine addiction in African Americans (allelic p=0.0086). For replication purposes, this SNP was analyzed in a larger independent population of cocaine addicted African Americans and controls and the association was confirmed (allelic p=4.53 × 10(-5); n=993). By performing a meta-analysis on the expanded populations, the statistical evidence for an association was substantially increased (allelic p=8.5 × 10(-7)) (p-values non-FDR corrected). CONCLUSION The present study suggests that polymorphisms in OPRD1 are relevant for cocaine addiction in the African American population and provides additional support for a broad role for OPRD1 variants in drug dependence.
Collapse
Affiliation(s)
- R C Crist
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Yoo JH, Kitchen I, Bailey A. The endogenous opioid system in cocaine addiction: what lessons have opioid peptide and receptor knockout mice taught us? Br J Pharmacol 2012; 166:1993-2014. [PMID: 22428846 DOI: 10.1111/j.1476-5381.2012.01952.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Cocaine addiction has become a major concern in the UK as Britain tops the European 'league table' for cocaine abuse. Despite its devastating health and socio-economic consequences, no effective pharmacotherapy for treating cocaine addiction is available. Identifying neurochemical changes induced by repeated drug exposure is critical not only for understanding the transition from recreational drug use towards compulsive drug abuse but also for the development of novel targets for the treatment of the disease and especially for relapse prevention. This article focuses on the effects of chronic cocaine exposure and withdrawal on each of the endogenous opioid peptides and receptors in rodent models. In addition, we review the studies that utilized opioid peptide or receptor knockout mice in order to identify and/or clarify the role of different components of the opioid system in cocaine-addictive behaviours and in cocaine-induced alterations of brain neurochemistry. The review of these studies indicates a region-specific activation of the µ-opioid receptor system following chronic cocaine exposure, which may contribute towards the rewarding effect of the drug and possibly towards cocaine craving during withdrawal followed by relapse. Cocaine also causes a region-specific activation of the κ-opioid receptor/dynorphin system, which may antagonize the rewarding effect of the drug, and at the same time, contribute to the stress-inducing properties of the drug and the triggering of relapse. These conclusions have important implications for the development of effective pharmacotherapy for the treatment of cocaine addiction and the prevention of relapse.
Collapse
Affiliation(s)
- Ji Hoon Yoo
- Division of Biochemistry, Faculty of Health & Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | | | | |
Collapse
|
50
|
Czoty PW, Nader MA. Individual differences in the effects of environmental stimuli on cocaine choice in socially housed male cynomolgus monkeys. Psychopharmacology (Berl) 2012; 224:69-79. [PMID: 22083591 PMCID: PMC3326438 DOI: 10.1007/s00213-011-2562-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 10/27/2011] [Indexed: 11/25/2022]
Abstract
RATIONALE Studies in laboratory animals have demonstrated an influence of environmentally derived stress and enrichment on the reinforcing effects of stimulants. OBJECTIVE To characterize the effects of acute exposure to ethologically valid environmental stimuli on the reinforcing strength of cocaine relative to food in socially housed monkeys. MATERIALS AND METHODS Choice between cocaine and food was assessed in subsets of 16 socially housed (4/pen) male cynomolgus monkeys immediately after the following manipulations: (1) treats placed in home cage, (2) a 10-min exposure to a rubber snake, or (3) 3 to 7 days of living in a larger environment without cage mates. RESULTS Placing treats in the home cage shifted the cocaine dose-response curve to the left in five monkeys tested and to the right in 4 of 12 animals. The rubber snake significantly shifted the cocaine choice curve to the left in dominant monkeys. Exposure to an enlarged environment decreased cocaine choice in 9 of 15 monkeys; this effect was transient and not related to social rank. Repeated testing did not affect cocaine choice. CONCLUSIONS Brief exposure to environmental events hypothesized to be stressors or enrichment altered cocaine choice, although not all individuals were affected and the effects were transient. Importantly, the data suggest that implementing positive changes in the environment produced effects that are clinically desirable. Understanding the behavioral and neurobiological mechanisms mediating sensitivity to environmental events in socially housed animals will lead to better treatment strategies for drug addiction.
Collapse
Affiliation(s)
- Paul W. Czoty
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Michael A. Nader
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|