1
|
Anghel SA, Dinu-Pirvu CE, Costache MA, Voiculescu AM, Ghica MV, Anuța V, Popa L. Receptor Pharmacogenomics: Deciphering Genetic Influence on Drug Response. Int J Mol Sci 2024; 25:9371. [PMID: 39273318 PMCID: PMC11395000 DOI: 10.3390/ijms25179371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
The paradigm "one drug fits all" or "one dose fits all" will soon be challenged by pharmacogenetics research and application. Drug response-efficacy or safety-depends on interindividual variability. The current clinical practice does not include genetic screening as a routine procedure and does not account for genetic variation. Patients with the same illness receive the same treatment, yielding different responses. Integrating pharmacogenomics in therapy would provide critical information about how a patient will respond to a certain drug. Worldwide, great efforts are being made to achieve a personalized therapy-based approach. Nevertheless, a global harmonized guideline is still needed. Plasma membrane proteins, like receptor tyrosine kinase (RTK) and G protein-coupled receptors (GPCRs), are ubiquitously expressed, being involved in a diverse array of physiopathological processes. Over 30% of drugs approved by the FDA target GPCRs, reflecting the importance of assessing the genetic variability among individuals who are treated with these drugs. Pharmacogenomics of transmembrane protein receptors is a dynamic field with profound implications for precision medicine. Understanding genetic variations in these receptors provides a framework for optimizing drug therapies, minimizing adverse reactions, and advancing the paradigm of personalized healthcare.
Collapse
Affiliation(s)
- Sorina Andreea Anghel
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Carol Davila", 6 Traian Vuia Str., 020956 Bucharest, Romania
- Department of Molecular Cell Biology, Institute of Biochemistry, Splaiul Independentei 296, 060031 Bucharest, Romania
| | - Cristina-Elena Dinu-Pirvu
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Carol Davila", 6 Traian Vuia Str., 020956 Bucharest, Romania
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), "Carol Davila" University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Mihaela-Andreea Costache
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Carol Davila", 6 Traian Vuia Str., 020956 Bucharest, Romania
| | - Ana Maria Voiculescu
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Carol Davila", 6 Traian Vuia Str., 020956 Bucharest, Romania
| | - Mihaela Violeta Ghica
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Carol Davila", 6 Traian Vuia Str., 020956 Bucharest, Romania
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), "Carol Davila" University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Valentina Anuța
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Carol Davila", 6 Traian Vuia Str., 020956 Bucharest, Romania
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), "Carol Davila" University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Lăcrămioara Popa
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Carol Davila", 6 Traian Vuia Str., 020956 Bucharest, Romania
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), "Carol Davila" University of Medicine and Pharmacy, 020956 Bucharest, Romania
| |
Collapse
|
2
|
Patrick C, Ettah U, Nguyen V, Hart C, Atchley E, Mallela K, Scheinman RI, Monte AA. Functional differences in the mu opioid receptor SNP 118A>G are dependent on receptor splice-variant and agonist-specific recruitment of β-arrestin. Clin Transl Sci 2024; 17:e13888. [PMID: 39118392 PMCID: PMC11310404 DOI: 10.1111/cts.13888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 08/10/2024] Open
Abstract
The OPRM1 gene codes for the mu opioid receptor (MOR) and polymorphisms are associated with complex patient clinical responses. The most studied single nucleotide polymorphism (SNP) in OPRM1 is adenine (A) substituted by guanine (G) at position 118 (118A>G, rs1799971) leading to a substitution of asparagine (Asn) for aspartic acid (Asp) at position 40 in the N terminus of the resulting protein. To date, no structural explanation for the associated clinical responses resulting from the 118A>G polymorphism has been proposed. We utilized computational modeling paired with functional cellular assays to predict unstructured N- and C-terminal regions of MOR-1. Using molecular docking and post-docking energy minimizations with morphine, we show that the extracellular substitution of Asn at position 40 alters the cytoplasmic C-terminal conformation, while leaving the G-protein binding interface unaffected. A real-time BRET assay measuring G-protein and β-arrestin association with MOR r generated data that tested this prediction. Consistent with this in silico prediction, we show changes in morphine-mediated β-arrestin association with receptor variants with little change in morphine-mediated G-protein association comparing MOR-1 wild type (WT) to MOR-1118A>G. We tested the system with different opioid agonists, the OPRM1 118A>G SNP, and different MOR splice variants (MOR-1 and MOR-1O). These results are consistent with the observation that patients with the 118A>G OPRM1 allele respond more readily to fentanyl than to morphine. In conclusion, the 118A>G substitution alters receptor responses to opioids through variable C-terminal domain movements that are agonist and splice variant dependent.
Collapse
Affiliation(s)
- Casey Patrick
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Utibeabasi Ettah
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Vu Nguyen
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Caitlin Hart
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Evan Atchley
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Krishna Mallela
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Robert I Scheinman
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Andrew A Monte
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
- Center for Bioinformatics & Personalized Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Rocky Mountain Poison & Drug Center, Denver Health and Hospital Authority, Denver, Colorado, USA
| |
Collapse
|
3
|
Ershad A, Shafiee Dolat Abadi S, Ebrahimian M, Hadeiy SK, Zamani N, Kolahi AA, Movafagh A, Hassanian-Moghaddam H. Association of the OPRM1 variant rs1799971 (A118G) and clinical manifestations in tramadol poisoned patients: a cross-sectional study. Clin Toxicol (Phila) 2024; 62:357-363. [PMID: 38946467 DOI: 10.1080/15563650.2024.2366921] [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: 07/12/2023] [Accepted: 06/06/2024] [Indexed: 07/02/2024]
Abstract
INTRODUCTION The opioid receptor mu1 is a protein coding gene that can have different codes for a protein and may have variations (polymorphisms) affecting how opioids work. The aim of this study was to investigate the prevalence of the most common opioid receptor mu1 polymorphism (A118G) and any relationship between this polymorphism and features following tramadol overdose. MATERIALS AND METHODS This was a cross-sectional study of patients admitted with tramadol poisoning to an Iranian hospital. These patients were not taking any other drugs or medications and had no history of seizures. RESULTS The results showed that among the 83 patients included in the study, 57 (69 per cent) had the AA genotype, 25 (30 per cent) had the AG genotype, and one (1 per cent) had the GG genotype for the opioid receptor mu1 A118G polymorphism. Nausea and/or vomiting occurred in nine (11 per cent) patients and dizziness in 38 (46 per cent) patients. Serious adverse events included seizures in 51 (60 per cent) patients and respiratory failure requiring mechanical ventilation in 21 (25 per cent) patients. However, there was no significant association between the opioid receptor mu1 A118G polymorphism and these adverse events. DISCUSSION In our study, the frequency of the A allele was greater than the G allele, and the AA genotype was more prevalent than AG. The GG genotype was the least common among the polymorphisms of opioid receptor mu1 rs1799971. There was no significant association between the opioid receptor mu1 A118G polymorphism and symptoms in tramadol-poisoned patients. Although these allele proportions are similar to the results reported in other Caucasian populations, they are dissimilar to the findings in Chinese and Singaporean populations. In these Asian studies, the predominant allele was the G allele. It has been suggested that a mutated G allele will decrease the production of opioid receptor mu1-related messenger ribonucleic acid and related proteins, leading to fewer mu-opioid receptors in the brain. CONCLUSIONS This study found no significant association between the opioid receptor mu1 A118G polymorphism and adverse outcomes in tramadol-poisoned patients. However, more research is needed to draw more definitive conclusions due to the limited evidence and variability of opioid receptor mu1 polymorphisms in different populations.
Collapse
Affiliation(s)
- Ali Ershad
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Seyed Kaveh Hadeiy
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Movafagh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Hassanian-Moghaddam
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Next Step Drug and Alcohol Services, Mental Health Commission, Perth, Australia
| |
Collapse
|
4
|
Cheong Y, Lee S, Okazawa H, Kosaka H, Jung M. Effects of functional polymorphisms of opioid receptor mu 1 and catechol-O-methyltransferase on the neural processing of pain. Psychiatry Clin Neurosci 2024; 78:300-308. [PMID: 38403942 PMCID: PMC11488594 DOI: 10.1111/pcn.13648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/14/2023] [Accepted: 01/18/2024] [Indexed: 02/27/2024]
Abstract
AIM Pain is reconstructed by brain activities and its subjectivity comes from an interplay of multiple factors. The current study aims to understand the contribution of genetic factors to the neural processing of pain. Focusing on the single-nucleotide polymorphism (SNP) of opioid receptor mu 1 (OPRM1) A118G (rs1799971) and catechol-O-methyltransferase (COMT) val158met (rs4680), we investigated how the two pain genes affect pain processing. METHOD We integrated a genetic approach with functional neuroimaging. We extracted genomic DNA information from saliva samples to genotype the SNP of OPRM1 and COMT. We used a percept-related model, in which two different levels of perceived pain intensities ("low pain: mildly painful" vs "high pain: severely painful") were employed as experimental stimuli. RESULTS Low pain involves a broader network relative to high pain. The distinct effects of pain genes were observed depending on the perceived pain intensity. The effects of low pain were found in supramarginal gyrus, angular gyrus, and anterior cingulate cortex (ACC) for OPRM1 and in middle temporal gyrus for COMT. For high pain, OPRM1 affected the insula and cerebellum, while COMT affected the middle occipital gyrus and ACC. CONCLUSION OPRM1 primarily affects sensory and cognitive components of pain processing, while COMT mainly influences emotional aspects of pain processing. The interaction of the two pain genes was associated with neural patterns coding for high pain and neural activation in the ACC in response to pain. The proteins encoded by the OPRM1 and COMT may contribute to the firing of pain-related neurons in the human ACC, a critical center for subjective pain experience.
Collapse
Grants
- 2022R1A6A3A01086118 National Research Foundation of Korea
- 2022R1F1A1066114 National Research Foundation of Korea
- 20H01766 Ministry of Education, Culture, Sports, Science and Technology
- 20H04272 Ministry of Education, Culture, Sports, Science and Technology
- 23-BR-04-03 Ministry of Science, ICT and Future Planning
- 23-BR-05-01 Ministry of Science, ICT and Future Planning
- 23-BR-04-03 Ministry of Science and ICT
- 23-BR-05-01 Ministry of Science and ICT
- National Research Foundation of Korea
- Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Yongjeon Cheong
- Cognitive Science Research GroupKorea Brain Research InstituteDaeguRepublic of Korea
| | - Seonkyoung Lee
- Cognitive Science Research GroupKorea Brain Research InstituteDaeguRepublic of Korea
| | - Hidehiko Okazawa
- Research Centre for Child Mental DevelopmentUniversity of FukuiEiheiji, FukuiJapan
- Division of Developmental Higher Brain Functions, Department of Child Development, United Graduate School of Child DevelopmentUniversity of FukuiFukuiJapan
| | - Hirotaka Kosaka
- Research Centre for Child Mental DevelopmentUniversity of FukuiEiheiji, FukuiJapan
- Division of Developmental Higher Brain Functions, Department of Child Development, United Graduate School of Child DevelopmentUniversity of FukuiFukuiJapan
- Department of NeuropsychiatryUniversity of FukuiFukuiJapan
| | - Minyoung Jung
- Cognitive Science Research GroupKorea Brain Research InstituteDaeguRepublic of Korea
| |
Collapse
|
5
|
Mills-Huffnagle SL, Morris C, Kawasaki S, Waverka GE, Hammen HA, Zgierska AE. First Response to Opioids Survey Tool (FROST): Pilot study of a brief screening tool to assess opioid use disorder risk. J Opioid Manag 2024; 20:197-207. [PMID: 39017612 DOI: 10.5055/jom.0860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
OBJECTIVE Early work suggests the type of subjective experiences upon first opioid use may predict opioid use disorder (OUD) risk. This study developed and pilot-tested a brief survey to evaluate the "first response" to opioids. DESIGN A cross-sectional survey research study. The survey was administered to a subsample for the second time to assess test-retest reliability. SETTING Outpatient. PARTICIPANTS Convenience sample of adults treated for OUD at an opioid treatment program. MAIN OUTCOME MEASURES A seven-question First Response to Opioids Survey Tool (FROST), developed based on the existing questionnaires and stakeholder-advisor feedback, was evaluated. RESULTS Participants (N = 157) were 36.8 (standard deviation [SD] = 9.4) years old, with 79.6 percent identifying as Caucasian and 56.7 percent male. They reported opioid initiation at 20.6 (SD = 8.8) years old, with a prescription-based (78.3 percent), orally administered (66.2 percent), and illicitly procured (51.0 percent) opioids. Upon opioid initiation, positive-valence, euphoria-like subjective experiences of feeling "comfortable" (65.0 percent), "happy" (61.1 percent), "euphoria" (58.6 percent), and "energized" (44.6 percent) were common, and different (p < 0.05) from other types of subjective experiences. Among 64 individuals who answered a question about "drug-liking," 50 (78.1 percent) reported drug-liking. Among 31 respondents who completed the survey a second time, the test-retest consistency was 78.2 percent for subjective experience characteristics and 72 percent for drug-liking responses. Qualitative results corroborated quantitative findings. CONCLUSION These results suggest that euphoria-type experiences and drug-liking upon opioid initiation are common among adults with OUD and FROST's promising psychometric properties. Future research should assess clinical utility of this brief survey, which could be applied at bedside and help identify those at risk for OUD, guide safer opioid prescribing, and reduce opioid-related harm.
Collapse
Affiliation(s)
- Sara L Mills-Huffnagle
- Penn State College of Medicine, Hershey, Pennsylvania. ORCID: https://orcid.org/0000-0002-6722-2586
| | | | | | | | | | | |
Collapse
|
6
|
Zhang Y, Randesi M, Blendy JA, Kreek MJ, Butelman ER. Impact of OPRM1 (Mu-opioid Receptor Gene) A112G Polymorphism on Dual Oxycodone and Cocaine Self-administration Behavior in a Mouse Model. Neuroscience 2024; 539:76-85. [PMID: 38211933 DOI: 10.1016/j.neuroscience.2024.01.002] [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: 03/31/2023] [Revised: 12/28/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024]
Abstract
The use of mu-opioid receptor (MOP-r) agonists such as oxycodone together with cocaine is prevalent, and deaths attributed to using these combinations have increased. RATIONALE It is unknown if functional single nucleotide polymorphisms (SNPs), such as the OPRM1 (MOP-r gene) SNP A118G, can predispose individuals to more dual opioid and psychostimulant intake. The dual self-administration (SA) of MOP-r agonists and cocaine has not been thoroughly examined, especially with regard to neurobiological changes. OBJECTIVES We examined oxycodone SA and subsequent dual oxycodone and cocaine SA in male and female A112G (A/G and G/G, heterozygote and homozygote, respectively) mice, models of human A118G carriers, versus wild-type (A/A) mice. METHODS Adult male and female A/G, G/G and A/A mice self-administered oxycodone (0.25 mg/kg/infusion, 4hr/session, FR 1.) for 10 consecutive days (sessions 1-10). Mice then self-administered cocaine (2 hr) following oxycodone SA (4 hr, as above) in each session for a further 10 consecutive days (sessions 11-20). Message RNA transcripts of 24 reward-related genes were examined in the dorsal striatum. RESULTS Male and female A/G and G/G mice had greater oxycodone SA than A/A mice did in the initial 10 days and in the last 10 sessions. Further, A/G and G/G mice showed greater cocaine intake than A/A mice. Dorsal striatal mRNA levels of Pdyn, Fkbp5, Oprk1, and Oprm1 were altered following oxycodone and cocaine SA. CONCLUSIONS These studies demonstrated that this functional genetic variation in Oprm1 affected dual opioid and cocaine SA and altered specific gene expression in the striatum.
Collapse
Affiliation(s)
- Yong Zhang
- Laboratory of the Biology of Addictive Diseases, the Rockefeller University, New York, NY 10065, United States.
| | - Matthew Randesi
- Laboratory of the Biology of Addictive Diseases, the Rockefeller University, New York, NY 10065, United States
| | - Julie A Blendy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, the Rockefeller University, New York, NY 10065, United States
| | - Eduardo R Butelman
- Laboratory of the Biology of Addictive Diseases, the Rockefeller University, New York, NY 10065, United States; Neuropsychoimaging of Addictions and Related Conditions Research Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| |
Collapse
|
7
|
Abstract
Major depressive disorder (MDD) is a leading cause of suicide in the world. Monoamine-based antidepressant drugs are a primary line of treatment for this mental disorder, although the delayed response and incomplete efficacy in some patients highlight the need for improved therapeutic approaches. Over the past two decades, ketamine has shown rapid onset with sustained (up to several days) antidepressant effects in patients whose MDD has not responded to conventional antidepressant drugs. Recent preclinical studies have started to elucidate the underlying mechanisms of ketamine's antidepressant properties. Herein, we describe and compare recent clinical and preclinical findings to provide a broad perspective of the relevant mechanisms for the antidepressant action of ketamine.
Collapse
Affiliation(s)
- Ji-Woon Kim
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
- College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Institute of Regulatory Innovation through Science, Kyung Hee University, Seoul, Republic of Korea
| | - Kanzo Suzuki
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Ege T Kavalali
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
| | - Lisa M Monteggia
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
| |
Collapse
|
8
|
Dunn KE, Huhn AS, Finan PH, Mange A, Bergeria CL, Maher BS, Rabinowitz JA, Strain EC, Antoine D. Polymorphisms in the A118G SNP of the OPRM1 gene produce different experiences of opioids: A human laboratory phenotype-genotype assessment. Addict Biol 2024; 29:e13355. [PMID: 38221808 PMCID: PMC10898793 DOI: 10.1111/adb.13355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/31/2023] [Accepted: 10/16/2023] [Indexed: 01/16/2024]
Abstract
Allelic variations in the A118G SNP of the OPRM1 gene change opioid signaling; however, evaluations of how allelic differences may influence opioid effects are lacking. This human laboratory paradigm examined whether the AA versus AG/GG genotypes determined opioid response profiles. Individuals with limited opioid exposure (N = 100) completed a five-day within-subject, double-blind, placebo-controlled, residential study. Participants were admitted (Day 1), received 4 mg hydromorphone (Day 2) and 0 mg, 2 mg and 8 mg hydromorphone in randomized order (Days 3-5) and completed self-reported visual analog scale (VAS) ratings and Likert scales, observed VAS, and physiological responses at baseline and for 6.5 h post-dose. Outcomes were analysed as peak/nadir effects over time as a function of genotype (available for N = 96 individuals; AG/GG = 13.5%, AA = 86.4%). Participants with AG/GG rated low and moderate doses of hydromorphone as significantly more positive (e.g., Good Effects VAS, coasting, drive, friendly, talkative, stimulation) with fewer negative effects (e.g., itchy skin, nausea, sleepiness), and were also observed as being more talkative and energetic relative to persons with AA. Persons with AG/GG were less physiologically reactive as determined by diastolic blood pressure and heart rate, but had more changes in core temperature compared with those with AA. Persons with AA also demonstrated more prototypic agonist effects across doses; persons with AG/GG showed limited response to 2 mg and 4 mg. Data suggest persons with AG/GG genotype experienced more pleasant and fewer unpleasant responses to hydromorphone relative to persons with AA. Future studies should replicate these laboratory findings in clinical populations to support a precision medicine approach to opioid prescribing.
Collapse
Affiliation(s)
- Kelly E. Dunn
- Department of Psychiatry and Behavioral SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Andrew S. Huhn
- Department of Psychiatry and Behavioral SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Patrick H. Finan
- Department of AnesthesiologyUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Ami Mange
- Yale School of MedicineNew HavenConnecticutUSA
| | - Cecilia L. Bergeria
- Department of Psychiatry and Behavioral SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Brion S. Maher
- Department of Mental HealthJohns Hopkins University School of Public HealthBaltimoreMarylandUSA
| | - Jill A. Rabinowitz
- Department of Mental HealthJohns Hopkins University School of Public HealthBaltimoreMarylandUSA
| | - Eric C. Strain
- Department of Psychiatry and Behavioral SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Denis Antoine
- Department of Psychiatry and Behavioral SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
9
|
Duffy EP, Bachtell RK, Ehringer MA. Opioid trail: Tracking contributions to opioid use disorder from host genetics to the gut microbiome. Neurosci Biobehav Rev 2024; 156:105487. [PMID: 38040073 PMCID: PMC10836641 DOI: 10.1016/j.neubiorev.2023.105487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023]
Abstract
Opioid use disorder (OUD) is a worldwide public health crisis with few effective treatment options. Traditional genetics and neuroscience approaches have provided knowledge about biological mechanisms that contribute to OUD-related phenotypes, but the complexity and magnitude of effects in the brain and body remain poorly understood. The gut-brain axis has emerged as a promising target for future therapeutics for several psychiatric conditions, so characterizing the relationship between host genetics and the gut microbiome in the context of OUD will be essential for development of novel treatments. In this review, we describe evidence that interactions between host genetics, the gut microbiome, and immune signaling likely play a key role in mediating opioid-related phenotypes. Studies in humans and model organisms consistently demonstrated that genetic background is a major determinant of gut microbiome composition. Furthermore, the gut microbiome is susceptible to environmental influences such as opioid exposure. Additional work focused on gene by microbiome interactions will be necessary to gain improved understanding of their effects on OUD-related behaviors.
Collapse
Affiliation(s)
- Eamonn P Duffy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA; Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA.
| | - Ryan K Bachtell
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Marissa A Ehringer
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA; Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
10
|
Takemura M, Niki K, Okamoto Y, Kawamura T, Kohno M, Matsuda Y, Ikeda K. Comparison of the Effects of OPRM1 A118G Polymorphism Using Different Opioids: A Prospective Study. J Pain Symptom Manage 2024; 67:39-49.e5. [PMID: 37757956 DOI: 10.1016/j.jpainsymman.2023.09.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/31/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023]
Abstract
CONTEXT μ-opioid receptor gene (OPRM1) A118G polymorphism (rs1799971) causes loss of N-glycosylation sites at the extracellular domain of μ-opioid receptors. G-allele carriers show a limited response to morphine; however, studies investigating the impact of A118G polymorphism on the efficacy of opioids other than morphine are limited. OBJECTIVE To compare the impact of A118G polymorphism on the efficacy of various opioids. METHODS This prospective cohort study enrolled 222 in-patients administered one of the following opioid therapies for cancer pain as part of an opioid introduction or rotation strategy: tapentadol extended-release tablets, methadone tablets, hydromorphone controlled-release tablets, oxycodone controlled-release tablets, or transdermal fentanyl patches. The impact of A118G polymorphism on the difference in the Brief Pain Inventory-Short Form score on days three, seven, and 14 from baseline was compared among the groups. RESULTS Overall, 81, 74, and 67 patients had the AA, AG, and GG genotypes, respectively, with an OPRM1 A118G G-allele variant frequency of 0.47. The reduction in the Brief Pain Inventory-Short Form score after opioid therapy initiation did not differ significantly among the patients with the three A118G genotypes treated with tapentadol (p = 0.84) or methadone (p = 0.97), whereas it was significantly smaller in G-allele carriers than that in AA homozygous patients treated with hydromorphone (p < 0.001), oxycodone (p = 0.031), or fentanyl (p < 0.001). CONCLUSION Tapentadol and methadone may be more suitable than hydromorphone, oxycodone, and fentanyl for G-allele carriers due to their dual mechanism of action and low susceptibility to OPRM1 A118G polymorphism.
Collapse
Affiliation(s)
- Miho Takemura
- Department of Clinical Pharmacy Research and Education (M.T., K.N., K.I.), Osaka University Graduate School of Pharmaceutical Sciences, Suita, Osaka, Japan; Department of Pharmacy (M.T., K.N., Y.O.), Ashiya Municipal Hospital, Ashiya, Hyogo, Japan.
| | - Kazuyuki Niki
- Department of Clinical Pharmacy Research and Education (M.T., K.N., K.I.), Osaka University Graduate School of Pharmaceutical Sciences, Suita, Osaka, Japan; Department of Pharmacy (M.T., K.N., Y.O.), Ashiya Municipal Hospital, Ashiya, Hyogo, Japan
| | - Yoshiaki Okamoto
- Department of Pharmacy (M.T., K.N., Y.O.), Ashiya Municipal Hospital, Ashiya, Hyogo, Japan
| | - Tomohiro Kawamura
- Department of Palliative Care (T.K., M.K., Y.M.), Ashiya Municipal Hospital, Ashiya, Hyogo, Japan
| | - Makie Kohno
- Department of Palliative Care (T.K., M.K., Y.M.), Ashiya Municipal Hospital, Ashiya, Hyogo, Japan
| | - Yoshinobu Matsuda
- Department of Palliative Care (T.K., M.K., Y.M.), Ashiya Municipal Hospital, Ashiya, Hyogo, Japan
| | - Kenji Ikeda
- Department of Clinical Pharmacy Research and Education (M.T., K.N., K.I.), Osaka University Graduate School of Pharmaceutical Sciences, Suita, Osaka, Japan
| |
Collapse
|
11
|
Frangakis SG, MacEachern M, Akbar TA, Bolton C, Lin V, Smith AV, Brummett CM, Bicket MC. Association of Genetic Variants with Postsurgical Pain: A Systematic Review and Meta-analyses. Anesthesiology 2023; 139:827-839. [PMID: 37774411 PMCID: PMC10859728 DOI: 10.1097/aln.0000000000004677] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2023]
Abstract
BACKGROUND Postsurgical pain is a key component of surgical recovery. However, the genetic drivers of postsurgical pain remain unclear. A broad review and meta-analyses of variants of interest will help investigators understand the potential effects of genetic variation. METHODS This article is a systematic review of genetic variants associated with postsurgical pain in humans, assessing association with postsurgical pain scores and opioid use in both acute (0 to 48 h postoperatively) and chronic (at least 3 months postoperatively) settings. PubMed, Embase, and the Cochrane Central Register of Controlled Trials were searched from 2000 to 2022 for studies using search terms related to genetic variants and postsurgical pain in humans. English-language studies in adult patients examining associations of one or more genetic variants with postsurgical pain were included. The primary outcome was association of genetic variants with either acute or chronic postsurgical pain. Pain was measured by patient-reported pain score or analgesic or opioid consumption. RESULTS A total of 163 studies were included, evaluating 129 unique genes and 594 unique genetic variants. Many of the reported significant associations fail to be replicated in other studies. Meta-analyses were performed for seven variants for which there was sufficient data (OPRM1 rs1799971; COMT rs4680, rs4818, rs4633, and rs6269; and ABCB1 rs1045642 and rs2032582). Only two variants were associated with small differences in postsurgical pain: OPRM1 rs1799971 (for acute postsurgical opioid use standard mean difference = 0.25; 95% CI, 0.16 to 0.35; cohort size, 8,227; acute postsurgical pain score standard mean difference = 0.20; 95% CI, 0.09 to 0.31; cohort size, 4,619) and COMT rs4680 (chronic postsurgical pain score standard mean difference = 0.26; 95% CI, 0.08 to 0.44; cohort size, 1,726). CONCLUSIONS Despite much published data, only two alleles have a small association with postsurgical pain. Small sample sizes, potential confounding variables, and inconsistent findings underscore the need to examine larger cohorts with consistent outcome measures. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Stephan G Frangakis
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Mark MacEachern
- Taubman Health Sciences Library, University of Michigan, Ann Arbor, Michigan
| | - T Adam Akbar
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, Michigan. Current Position: Department of Anesthesiology, Northwestern Medicine, Chicago, Illinois
| | - Christian Bolton
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Victor Lin
- Victor Lin, D.O., Ph.D.; Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Albert V Smith
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Chad M Brummett
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, Michigan; Opioid Prescribing and Engagement Network, Institute for Healthcare Innovation and Policy, University of Michigan, Ann Arbor, Michigan
| | - Mark C Bicket
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, Michigan; Opioid Prescribing and Engagement Network, Institute for Healthcare Innovation and Policy, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
12
|
Kong L, Wang J, Guan S, Chen X, Li M, Gao L, Zhong D, Zhang L. Nomogram for predicting opioid-induced nausea and vomiting for cancer pain patients. Support Care Cancer 2023; 31:663. [PMID: 37914831 PMCID: PMC10620250 DOI: 10.1007/s00520-023-08144-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023]
Abstract
OBJECTIVE Opioid-induced nausea and vomiting are frequently observed as an adverse effect in the treatment of cancer-related pain. The factors that affect OINV in cancer patients remain unclear. In this study, we developed a nomogram for predicting the occurrence of OINV in this population using retrospective clinical data. METHODS We collected data from 416 cancer pain patients, 70% of whom used the training set to analyze demographic and clinical variables. We used multivariate logistic regression to identify significant factors associated with OINV. Then, we construct a prediction nomogram. The validation set comprises the remaining 30%. The reliability of the nomogram is evaluated by bootstrap resampling. RESULTS Using multivariate logistic regression, we identified five significant factors associated with OINV. The C-index was 0.835 (95% confidence interval [CI], 0.828-0.842) for the training set and 0.810 (95% CI, 0.793-0.826) for the validation set. The calibrated curves show a good agreement between the predicted and actual occurrence of OINV. CONCLUSION In a retrospective study based on five saliency-found variables, we developed and proved a reliable nomogram model to predict OINV in cancer pain patients. Future prospective studies should assess the model's reliability and usefulness in clinical practice.
Collapse
Affiliation(s)
- Lingping Kong
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jing Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Shasha Guan
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaochen Chen
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Meiqing Li
- Department of Oncology, People's Hospital of Inner Mongolia Autonomous Region, Hohhot, 010017, Inner Mongolia, China
| | - Liming Gao
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, 066000, Hebei, China
| | - Diansheng Zhong
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Linlin Zhang
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
13
|
Gameiro‐Ros I, Popova D, Prytkova I, Pang ZP, Liu Y, Dick D, Bucholz KK, Agrawal A, Porjesz B, Goate AM, Xuei X, Kamarajan C, Tischfield JA, Edenberg HJ, Slesinger PA, Hart RP. 5. Collaborative Study on the Genetics of Alcoholism: Functional genomics. GENES, BRAIN, AND BEHAVIOR 2023; 22:e12855. [PMID: 37533187 PMCID: PMC10550792 DOI: 10.1111/gbb.12855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/31/2023] [Accepted: 06/17/2023] [Indexed: 08/04/2023]
Abstract
Alcohol Use Disorder is a complex genetic disorder, involving genetic, neural, and environmental factors, and their interactions. The Collaborative Study on the Genetics of Alcoholism (COGA) has been investigating these factors and identified putative alcohol use disorder risk genes through genome-wide association studies. In this review, we describe advances made by COGA in elucidating the functional changes induced by alcohol use disorder risk genes using multimodal approaches with human cell lines and brain tissue. These studies involve investigating gene regulation in lymphoblastoid cells from COGA participants and in post-mortem brain tissues. High throughput reporter assays are being used to identify single nucleotide polymorphisms in which alternate alleles differ in driving gene expression. Specific single nucleotide polymorphisms (both coding or noncoding) have been modeled using induced pluripotent stem cells derived from COGA participants to evaluate the effects of genetic variants on transcriptomics, neuronal excitability, synaptic physiology, and the response to ethanol in human neurons from individuals with and without alcohol use disorder. We provide a perspective on future studies, such as using polygenic risk scores and populations of induced pluripotent stem cell-derived neurons to identify signaling pathways related with responses to alcohol. Starting with genes or loci associated with alcohol use disorder, COGA has demonstrated that integration of multimodal data within COGA participants and functional studies can reveal mechanisms linking genomic variants with alcohol use disorder, and potential targets for future treatments.
Collapse
Affiliation(s)
- Isabel Gameiro‐Ros
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Dina Popova
- Human Genetics Institute of New JerseyRutgers UniversityPiscatawayNew JerseyUSA
| | - Iya Prytkova
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Zhiping P. Pang
- Human Genetics Institute of New JerseyRutgers UniversityPiscatawayNew JerseyUSA
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical SchoolRutgers UniversityNew BrunswickNew JerseyUSA
| | - Yunlong Liu
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Danielle Dick
- Rutgers Addiction Research Center, Robert Wood Johnson Medical SchoolRutgers UniversityPiscatawayNew JerseyUSA
| | - Kathleen K. Bucholz
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Arpana Agrawal
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Bernice Porjesz
- Department of Psychiatry and Behavioral SciencesSUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Alison M. Goate
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Xiaoling Xuei
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Chella Kamarajan
- Department of Psychiatry and Behavioral SciencesSUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | | | - Jay A. Tischfield
- Human Genetics Institute of New JerseyRutgers UniversityPiscatawayNew JerseyUSA
- Department of GeneticsRutgers UniversityPiscatawayNew JerseyUSA
| | - Howard J. Edenberg
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana UniversityIndianapolisIndianaUSA
| | - Paul A. Slesinger
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ronald P. Hart
- Human Genetics Institute of New JerseyRutgers UniversityPiscatawayNew JerseyUSA
- Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNew JerseyUSA
| |
Collapse
|
14
|
Blum K, Gold MS, Cadet JL, Gondre-Lewis MC, McLaughlin T, Braverman ER, Elman I, Paul Carney B, Cortese R, Abijo T, Bagchi D, Giordano J, Dennen CA, Baron D, Thanos PK, Soni D, Makale MT, Makale M, Murphy KT, Jafari N, Sunder K, Zeine F, Ceccanti M, Bowirrat A, Badgaiyan RD. Invited Expert Opinion- Bioinformatic and Limitation Directives to Help Adopt Genetic Addiction Risk Screening and Identify Preaddictive Reward Dysregulation: Required Analytic Evidence to Induce Dopamine Homeostatsis. MEDICAL RESEARCH ARCHIVES 2023; 11:10.18103/mra.v11i8.4211. [PMID: 37885438 PMCID: PMC10601302 DOI: 10.18103/mra.v11i8.4211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Addiction, albeit some disbelievers like Mark Lewis [1], is a chronic, relapsing brain disease, resulting in unwanted loss of control over both substance and non- substance behavioral addictions leading to serious adverse consequences [2]. Addiction scientists and clinicians face an incredible challenge in combatting the current opioid and alcohol use disorder (AUD) pandemic throughout the world. Provisional data from the Centers for Disease Control and Prevention (CDC) shows that from July 2021-2022, over 100,000 individuals living in the United States (US) died from a drug overdose, and 77,237 of those deaths were related to opioid use [3]. This number is expected to rise, and according to the US Surgeon General it is highly conceivable that by 2025 approximately 165,000 Americans will die from an opioid overdose. Alcohol abuse, according to data from the World Health Organization (WHO), results in 3 million deaths worldwide every year, which represents 5.3% of all deaths globally [4].
Collapse
Affiliation(s)
- Kenneth Blum
- The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX., USA
- Division of Addiction Research & Education, Center for Sports, Exercise & Psychiatry, Western University Health Sciences, Pomona, CA., USA
- Institute of Psychology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Psychiatry, School of Medicine, University of Vermont, Burlington, VT.,USA
- Department of Psychiatry, Wright State University Boonshoft School of Medicine and Dayton VA Medical Centre, Dayton, OH, USA
- Division of Nutrigenomics Research, TranspliceGen Therapeutics, Inc., Austin, Tx., 78701, USA
- Department of Nutrigenomic Research, Victory Nutrition International, Inc., Bonita Springs, FL, USA
- Division of Personalized Medicine, Cross-Cultural Research and Educational Institute, San Clemente, CA., USA
- Sunder Foundation, Palm Springs, CA, USA
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Mark S Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO., USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD., USA
| | - Marjorie C. Gondre-Lewis
- Neuropsychopharmacology Laboratory, Department of Anatomy, Howard University College of Medicine, Washington, DC., USA
| | - Thomas McLaughlin
- Division of Nutrigenomics Research, TranspliceGen Therapeutics, Inc., Austin, Tx., 78701, USA
| | - Eric R Braverman
- The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX., USA
| | - Igor Elman
- Center for Pain and the Brain (P.A.I.N Group), Department of Anesthesiology, Critical Care & Pain Medicine, Boston Children’s Hospital, Boston, MA., USA
| | - B. Paul Carney
- Division Pediatric Neurology, University of Missouri, School of Medicine, Columbia, MO., USA
| | - Rene Cortese
- Department of Child Health – Child Health Research Institute, & Department of Obstetrics, Gynecology and Women’s Health School of Medicine, University of Missouri, MO., USA
| | - Tomilowo Abijo
- Neuropsychopharmacology Laboratory, Department of Anatomy, Howard University College of Medicine, Washington, DC., USA
| | - Debasis Bagchi
- Department of Pharmaceutical Sciences, Texas Southern University College of Pharmacy and Health Sciences, Houston, TX, USA
| | - John Giordano
- Division of Personalized Mental Illness Treatment & Research, Ketamine Infusion Clinics of South Florida, Pompano Beach, Fl., USA
| | - Catherine A. Dennen
- Department of Family Medicine, Jefferson Health Northeast, Philadelphia, PA, USA
| | - David Baron
- Institute of Psychology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Diwanshu Soni
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA., USA
| | - Milan T. Makale
- Department of Radiation Medicine and Applied Sciences, UC San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, USA
| | - Miles Makale
- Department of Psychology, UC San Diego, Health Sciences Drive, La Jolla, CA, 92093, USA
| | | | - Nicole Jafari
- Department of Human Development, California State University at long Beach, Long Beach, CA., USA
- Division of Personalized Medicine, Cross-Cultural Research and Educational Institute, San Clemente, CA., USA
| | - Keerthy Sunder
- Department of Psychiatry, Menifee Global Medical Center, Palm Desert, CA., USA
- Sunder Foundation, Palm Springs, CA, USA
| | - Foojan Zeine
- Awareness Integration Institute, San Clemente, CA., USA
- Department of Health Science, California State University at Long Beach, Long Beach, CA., USA
| | - Mauro Ceccanti
- Società Italiana per il Trattamento dell’Alcolismo e le sue Complicanze (SITAC), ASL Roma1, Sapienza University of Rome, Rome, Italy
| | - Abdalla Bowirrat
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Rajendra D. Badgaiyan
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, Long School of Medicine, University of Texas Medical Center, San Antonio, TX., USA
- Department of Psychiatry, Mt Sinai University School of Medicine, New York, NY., USA
| |
Collapse
|
15
|
Bagher AM, Hareeri RH. Allele frequency and genotype distribution of the opioid receptor μ-1 (OPRM1) A118G polymorphism in the Western Saudi population. J Appl Biomed 2023; 21:160-165. [PMID: 37747315 DOI: 10.32725/jab.2023.012] [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: 10/25/2022] [Accepted: 09/14/2023] [Indexed: 09/26/2023] Open
Abstract
The single nucleotide polymorphism (SNP) A118G (rs1799971) in the Mu Opioid Receptor 1 (OPRM1) gene is associated with significant variations in analgesic doses and adverse effects of opioids. The A118G OPRM1 allele distributions vary significantly between different populations worldwide. The study aimed to assess the allele frequency and genotype distribution of OPRM1 A118G SNP in Saudis. This cross-sectional study included 124 healthy Saudis (62 males and 62 females) visiting the King Abdulaziz University Hospital in Jeddah, Saudi Arabia. The Oragene®-DISCOVER (OGR-600) kits were used to collect saliva samples from the participants. Polymerase chain reaction-restriction fragment length polymorphism was utilized to assess the SNP. Among the tested population, 79.03% (95% C.I. 70.81-85.82) were homozygous wild-type A118A, 16.13% (95% C.I. 10.14-23.80) were heterozygous A118G, and 4.84% (95% C.I. 1.80-10.23) were homozygous mutant G118G. OPRM1 A118G polymorphism allele frequencies were 87% (95% C.I. 79.89-92.44) and 13% (95% C.I. 7.56-20.11) for the 118A and 118G alleles, respectively. A higher frequency of the OPRM1 118G allele was present in females, 21% (95% C.I. 11.66-33.17) compared to males, 5% (95% C.I. 1.01-13.50). Relative to other Asian countries, the Saudi population showed a low prevalence of the OPRM1 A118G polymorphism, with a higher frequency of the 118G allele in females. Our research will contribute to the existing knowledge on the prevalence of OPRM1 A118G polymorphism, which could be considered for the personalized prescribing of opioid analgesics.
Collapse
Affiliation(s)
- Amina M Bagher
- King AbdulAziz University, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Jeddah, Saudi Arabia
| | - Rawan H Hareeri
- King AbdulAziz University, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Jeddah, Saudi Arabia
| |
Collapse
|
16
|
Suresh N, Kantipudi SJ, Ramu D, Muniratnam SK, Venkatesan V. Association between opioid and dopamine receptor gene polymorphisms OPRM1 rs1799971, DAT VNTR 9-10 repeat allele, DRD1 rs4532 and DRD2 rs1799732 and alcohol dependence: an ethnicity oriented meta-analysis. Pharmacogenet Genomics 2023; 33:139-152. [PMID: 37466123 DOI: 10.1097/fpc.0000000000000502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
OBJECTIVE We carried out a meta-analysis of four opioid and dopamine candidate gene polymorphisms having conflicting results in prior literature, namely OPRM1 rs1799971, DAT VNTR 9-10 repeat, DRD1 rs4532 and DRD2 rs1799732, to clarify their association with alcohol dependence and further stratified results by ethnicity to analyze possible ethnicity-mediated effects. METHODS Inclusion criteria: case-control studies assessing the association between OPRM1 rs1799971, DAT VNTR 9/10 repeat allele, DRD1 rs4532 and DRD2 rs1799732 with alcohol dependence, with sufficient data available to calculate the odds ratio (OR) within a 95% confidence interval. Exclusion criteria: studies of quantitative measures of alcohol consumption, response to medications or analyses of other markers in the candidate genes, studies without controls, animal studies and lack of genotyping data. Information sources were PubMed, Google Scholar and ScienceDirect databases, all of which were searched for articles published till 2021. Heterogeneity between studies and publication bias, subgroup analyses and sensitivity analyses were carried out. RESULTS A total of 41 published studies were included in the current meta-analysis. For the OPRM1 gene, there was a statistically significant association in the Asian population with a pooled OR of 1.707 (95% CI, 1.32-2.20 P < 0.0001) and 1.618 (95% CI, 1.16-2.26 P = 0.005) in the additive and dominant genetic models. For DAT VNTR 9/10 repeat, a statistically significant association of the risk vs. common allele was observed in AD with a pooled OR of 1.104 (95% CI, 1.00-1.21 P = 0.046) in the allele model and the additive genetic model in the Caucasian population with pooled OR of 1.152 (95% CI, 1.01-1.31 P = 0.034). CONCLUSION Results indicate that some of the effects may be ethnicity-specific. OTHER The meta-analysis has been registered in the CRD PROSPERO (CRD42023411576).
Collapse
Affiliation(s)
| | | | - Deepika Ramu
- Department of Human Genetics SRIHER, Porur, Chennai
| | | | | |
Collapse
|
17
|
Kumar S, Kesavan R, Sistla SC, Penumadu P, Natarajan H, Nair S, Chakradhara Rao US, Venkatesan V, Kundra P. Impact of Genetic Variants on Postoperative Pain and Fentanyl Dose Requirement in Patients Undergoing Major Breast Surgery: A Candidate Gene Association Study. Anesth Analg 2023; 137:409-417. [PMID: 36538471 DOI: 10.1213/ane.0000000000006330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Postoperative analgesia is crucial for the early and effective recovery of patients undergoing surgery. Although postoperative multimodal analgesia is widely practiced, opioids such as fentanyl are still one of the best analgesics. The analgesic response of fentanyl varies widely among individuals, probably due to genetic and nongenetic factors. Among genetic factors, single nucleotide polymorphisms (SNPs) may influence its analgesic response by altering the structure or function of genes involved in nociceptive, fentanyl pharmacodynamic, and pharmacokinetic pathways. Thus, it is necessary to comprehensively ascertain if the SNPs present in the aforementioned pathways are associated with interindividual differences in fentanyl requirement. In this study, we evaluated the association between 10 candidate SNPs in 9 genes and 24-hour postoperative fentanyl dose (primary outcome) and also with postoperative pain scores and time for first analgesia (secondary outcomes). METHODS A total of 257 South Indian women, aged 18-70 years, with American Society of Anesthesiologists (ASA) physical status I-III, undergoing major breast surgery under general anesthesia, were included in the study. Patients were genotyped for candidate SNPs using real-time polymerase chain reaction. All patients received a standardized intravenous fentanyl infusion through a patient-controlled analgesic (PCA) pump, and the 24-hour postoperative fentanyl dose requirement was measured using PCA. RESULTS The median 24-hour postoperative fentanyl requirement was higher in rs1799971 carriers (G/G versus A/A + A/G-620 μg [500-700] vs 460 μg [400-580]) with a geometric mean (GM) ratio of 1.91 (95% confidence interval [CI], 1.071-1.327). The median 24-hour pain scores were higher in rs4680 carriers (A/G + A/A versus G/G-34 [30-38] vs 31 [30-38]) with a GM ratio of 1.059 (95% CI, 1.018-1.101) and were lower in rs1045642 carriers (A/A + A/G versus G/G-34 [30-38] vs 30 [30-34]) with a GM ratio of 0.936 (95% CI, 0.889-0.987). The median time for first analgesic was lower in rs734784 carriers [C/C versus T/T + C/T-240 minutes (180-270) vs 240 minutes (210-270)] with a GM ratio of 0.902 (95% CI, 0.837-0.972). Five of 9 clinical factors, namely, history of diabetes, hypertension, hypothyroidism, anesthesia duration, and intraoperative fentanyl requirement were associated with different outcomes individually ( P < .05) and were used to adjust the respective associations. CONCLUSIONS The SNP opioid receptor mu-1 ( OPRM1 ) (rs1799971) was associated with higher postoperative fentanyl requirement in South Indian patients undergoing major breast surgery. Twenty-four hour postoperative pain scores were higher in catechol-O-methyl transferase ( COMT ) (rs4680) carriers and lower in ATP binding cassette subfamily B member 1 ( ABCB1 ) (rs1045642) carriers, whereas time for first analgesic was lower in potassium channel subunit 1 ( KCNS1 ) (rs734784) carriers. However, these exploratory findings must be confirmed in a larger study.
Collapse
Affiliation(s)
- Shathish Kumar
- From the Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Ramasamy Kesavan
- From the Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Sarath Chandra Sistla
- Department of General Surgery, Sri Manakula Vinayagar Medical College and Hospital, Puducherry, India; Departments of
| | | | - Harivenkatesh Natarajan
- From the Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | | | - Uppugunduri S Chakradhara Rao
- Faculty of Medicine, CANSEARCH Research Platform in Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
| | - Vasuki Venkatesan
- Indian Council of Medical Research-Vector Control Research Centre, Department of Health Research, Ministry of Health & Family Welfare, GOI, Puducherry, India
| | - Pankaj Kundra
- Department of Anaesthesiology, JIPMER, Puducherry, India
| |
Collapse
|
18
|
Agulló L, Aguado I, Muriel J, Margarit C, Gómez A, Escorial M, Sánchez A, Fernández A, Peiró AM. Pharmacogenetic Guided Opioid Therapy Improves Chronic Pain Outcomes and Comorbid Mental Health: A Randomized, Double-Blind, Controlled Study. Int J Mol Sci 2023; 24:10754. [PMID: 37445931 DOI: 10.3390/ijms241310754] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/20/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Interindividual variability in analgesic response is at least partly due to well-characterized polymorphisms that are associated with opioid dosing and adverse outcomes. The Clinical Pharmacogenetics Implementation Consortium (CPIC) has put forward recommendations for the CYP2D6 phenotype, but the list of studied drug-gene pairs continues to grow. This clinical trial randomized chronic pain patients (n = 60), referred from primary care to pain unit care into two opioid prescribing arms, one guided by CYP2D6, μ-opioid receptor (OPRM1), and catechol-O-methyl transferase (COMT) genotypes vs. one with clinical routine. The genotype-guided treatment reduced pain intensity (76 vs. 59 mm, p < 0.01) by improving pain relief (28 vs. 48 mm, p < 0.05), increased quality of life (43 vs. 56 mm p < 0.001), and lowered the incidence of clinically relevant adverse events (3 [1-5] vs. 1 [0-2], p < 0.01) and 42% opioid dose (35 [22-61] vs. 60 [40-80] mg/day, p < 0.05) as opposed to usual prescribing arm. The final health utility score was significantly higher (0.71 [0.58-0.82] vs. 0.51 [0.13-0.67] controls, p < 0.05) by improving sleepiness and depression comorbidity, with a significant reduction of 30-34% for headache, dry mouth, nervousness, and constipation. A large-scale implementation analysis could help clinical translation, together with a pharmaco-economic evaluation.
Collapse
Affiliation(s)
- Laura Agulló
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Isidro Aguado
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
| | - Javier Muriel
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - César Margarit
- Pain Unit, Department of Health of Alicante, General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
| | - Alba Gómez
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Mónica Escorial
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Astrid Sánchez
- San Vicente del Raspeig II Health Center, c/Alicante, 78, Sant Vicent del Raspeig, 03690 Alicante, Spain
| | - Alicia Fernández
- San Vicente del Raspeig II Health Center, c/Alicante, 78, Sant Vicent del Raspeig, 03690 Alicante, Spain
| | - Ana M Peiró
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), General University Hospital of Alicante, c/Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Pharmacology, Toxicology and Chemical Safety Unit, Institute of Bioengineering, Miguel Hernández University, Avda. de la Universidad s/n, 03202 Elche, Spain
| |
Collapse
|
19
|
Diwan AD, Melrose J. Intervertebral disc degeneration and how it leads to low back pain. JOR Spine 2023; 6:e1231. [PMID: 36994466 PMCID: PMC10041390 DOI: 10.1002/jsp2.1231] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 09/23/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
The purpose of this review was to evaluate data generated by animal models of intervertebral disc (IVD) degeneration published in the last decade and show how this has made invaluable contributions to the identification of molecular events occurring in and contributing to pain generation. IVD degeneration and associated spinal pain is a complex multifactorial process, its complexity poses difficulties in the selection of the most appropriate therapeutic target to focus on of many potential candidates in the formulation of strategies to alleviate pain perception and to effect disc repair and regeneration and the prevention of associated neuropathic and nociceptive pain. Nerve ingrowth and increased numbers of nociceptors and mechanoreceptors in the degenerate IVD are mechanically stimulated in the biomechanically incompetent abnormally loaded degenerate IVD leading to increased generation of low back pain. Maintenance of a healthy IVD is, thus, an important preventative measure that warrants further investigation to preclude the generation of low back pain. Recent studies with growth and differentiation factor 6 in IVD puncture and multi-level IVD degeneration models and a rat xenograft radiculopathy pain model have shown it has considerable potential in the prevention of further deterioration in degenerate IVDs, has regenerative properties that promote recovery of normal IVD architectural functional organization and inhibits the generation of inflammatory mediators that lead to disc degeneration and the generation of low back pain. Human clinical trials are warranted and eagerly anticipated with this compound to assess its efficacy in the treatment of IVD degeneration and the prevention of the generation of low back pain.
Collapse
Affiliation(s)
- Ashish D. Diwan
- Spine Service, Department of Orthopaedic Surgery, St. George & Sutherland Clinical SchoolUniversity of New South WalesSydneyNew South WalesAustralia
| | - James Melrose
- Raymond Purves Bone and Joint Research LaboratoryKolling Institute, Sydney University Faculty of Medicine and Health, Northern Sydney Area Health District, Royal North Shore HospitalSydneyNew South WalesAustralia
- Graduate School of Biomedical EngineeringThe University of New South WalesSydneyNew South WalesAustralia
| |
Collapse
|
20
|
Margolis EB, Moulton MG, Lambeth PS, O'Meara MJ. The life and times of endogenous opioid peptides: Updated understanding of synthesis, spatiotemporal dynamics, and the clinical impact in alcohol use disorder. Neuropharmacology 2023; 225:109376. [PMID: 36516892 PMCID: PMC10548835 DOI: 10.1016/j.neuropharm.2022.109376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The opioid G-protein coupled receptors (GPCRs) strongly modulate many of the central nervous system structures that contribute to neurological and psychiatric disorders including pain, major depressive disorder, and substance use disorders. To better treat these and related diseases, it is essential to understand the signaling of their endogenous ligands. In this review, we focus on what is known and unknown about the regulation of the over two dozen endogenous peptides with high affinity for one or more of the opioid receptors. We briefly describe which peptides are produced, with a particular focus on the recently proposed possible synthesis pathways for the endomorphins. Next, we describe examples of endogenous opioid peptide expression organization in several neural circuits and how they appear to be released from specific neural compartments that vary across brain regions. We discuss current knowledge regarding the strength of neural activity required to drive endogenous opioid peptide release, clues about how far peptides diffuse from release sites, and their extracellular lifetime after release. Finally, as a translational example, we discuss the mechanisms of action of naltrexone (NTX), which is used clinically to treat alcohol use disorder. NTX is a synthetic morphine analog that non-specifically antagonizes the action of most endogenous opioid peptides developed in the 1960s and FDA approved in the 1980s. We review recent studies clarifying the precise endogenous activity that NTX prevents. Together, the works described here highlight the challenges and opportunities the complex opioid system presents as a therapeutic target.
Collapse
Affiliation(s)
- Elyssa B Margolis
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, CA, USA.
| | - Madelyn G Moulton
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Philip S Lambeth
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew J O'Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
21
|
Khani P, Ansari Dezfouli M, Nasri F, Rahemi M, Ahmadloo S, Afkhami H, Saeidi F, Tereshchenko S, Bigdeli MR, Modarressi MH. Genetic and epigenetic effects on couple adjustment in context of romantic relationship: A scoping systematic review. Front Genet 2023; 14:1002048. [PMID: 36816018 PMCID: PMC9937082 DOI: 10.3389/fgene.2023.1002048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 01/02/2023] [Indexed: 01/26/2023] Open
Abstract
Introduction: Couples' relationships defined by a complex interaction between the two partners and their intrapersonal traits. Romantic; relationships and love are associated with marital satisfaction and stability, as well as couples' happiness and health. Personality traits influence romantic relationships and, personality influenced by genetical and non-genetically factors. The roles of non-genetically factors such as socioeconomic position and external appearance have revealed in determining the quality of romantic relationships. Methods: We; performed a scoping systematic review to assess the association between genetics and epigenetic factors and romantic relationship. Relevant articles were identified by PubMed, EMBASE, Web of Science, Scopus, and the APA PsycInfo searching between inception and 4 June 2022. Results: Different studies evaluated the associated polymorphisms in 15 different genes or chromosomal regions. In the first step; we classified them into four groups: (1) Oxytocin-related signaling pathway (OXTR, CD38, and AVPR1A); (2) Serotonin-related signaling pathway (SLC6A4, HTR1A, and HTR2A); (3) Dopamine and catecholamine-related signaling pathway (DRD1, DRD2, DRD4, ANKK1, and COMT); and (4) other genes (HLA, GABRA2, OPRM1, and Y-DNA haplogroup D-M55). Then, we evaluated and extracted significant polymorphisms that affect couple adjustment and romantic relationships. Discussion: Overall, the findings suggest that genetic and epigenetics variants play a key role in marital adjustment and romantic relationships over time.
Collapse
Affiliation(s)
- Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mitra Ansari Dezfouli
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Farzad Nasri
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Rahemi
- Department of stem cell technology and tissue regeneration, Faculty of Science, Tehran University, Tehran, Iran
| | - Salma Ahmadloo
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran,Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran
| | - Hamed Afkhami
- Department of Medical Microbiology, Faculty of Medicine, Shahed University of Medical Sciences, Tehran, Iran
| | - Farzane Saeidi
- Department of Medical Genetics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sergey Tereshchenko
- Research Institute of Medical Problems of the North, Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences”, Krasnoyarsk, Russia,*Correspondence: Sergey Tereshchenko, ; Mohammad Reza Bigdeli, ; Mohammad Hossein Modarressi,
| | - Mohammad Reza Bigdeli
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran,Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran,*Correspondence: Sergey Tereshchenko, ; Mohammad Reza Bigdeli, ; Mohammad Hossein Modarressi,
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran,*Correspondence: Sergey Tereshchenko, ; Mohammad Reza Bigdeli, ; Mohammad Hossein Modarressi,
| |
Collapse
|
22
|
Noufal Y, Kringel D, Toennes SW, Dudziak R, Lötsch J. Pharmacological data science perspective on fatal incidents of morphine treatment. Pharmacol Ther 2023; 241:108312. [PMID: 36423714 DOI: 10.1016/j.pharmthera.2022.108312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022]
Abstract
Morphine prescribed for analgesia has caused drug-related deaths at an estimated incidence of 0.3% to 4%. Morphine has pharmacological properties that make it particularly difficult to assess the causality of morphine administration with a patient's death, such as its slow transfer between plasma and central nervous sites of action and the existence of the active metabolite morphine-6-glucuronide with opioid agonistic effects, Furthermore, there is no well-defined toxic dose or plasma/blood concentration for morphine. Dosing is often adjusted for adequate pain relief. Here, we summarize reported deaths associated with morphine therapy, including associated morphine exposure and modulating patient factors such as pharmacogenetics, concomitant medications, or comorbidities. In addition, we systematically analyzed published numerical information on the stability of concentrations of morphine and its relevant metabolites in biological samples collected postmortem. A medicolegal case is presented in which the causality of morphine administration with death was in dispute and pharmacokinetic modeling was applied to infer the administered dose. The results of this analytical review suggest that (i) inference from postmortem blood concentrations to the morphine dose administered has low validity and (ii) causality between a patient's death and the morphine dose administered remains a highly context-dependent and collaborative assessment among experts from different medical specialties.
Collapse
Affiliation(s)
- Yazan Noufal
- Goethe-University, Institute of Clinical Pharmacology, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Dario Kringel
- Goethe-University, Institute of Clinical Pharmacology, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Stefan W Toennes
- Goethe-University, University Hospital Frankfurt, Institute of Legal Medicine, Kennedyallee 104, 60596 Frankfurt am Main, Germany
| | - Rafael Dudziak
- Goethe-University, University Hospital Frankfurt, Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jörn Lötsch
- Goethe-University, Institute of Clinical Pharmacology, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.
| |
Collapse
|
23
|
Tour J, Sandström A, Kadetoff D, Schalling M, Kosek E. The OPRM1 gene and interactions with the 5-HT1a gene regulate conditioned pain modulation in fibromyalgia patients and healthy controls. PLoS One 2022; 17:e0277427. [PMID: 36342939 PMCID: PMC9639841 DOI: 10.1371/journal.pone.0277427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022] Open
Abstract
Fibromyalgia (FM) patients have dysfunctional endogenous pain modulation, where opioid and serotonergic signaling is implicated. The aim of this study was to investigate whether genetic variants in the genes coding for major structures in the opioid and serotonergic systems can affect pain modulation in FM patients and healthy controls (HC). Conditioned pain modulation (CPM), evaluating the effects of ischemic pain on pressure pain sensitivity, was performed in 82 FM patients and 43 HC. All subjects were genotyped for relevant functional polymorphisms in the genes coding for the μ-opioid receptor (OPRM1, rs1799971), the serotonin transporter (5-HTT, 5-HTTLPR/rs25531) and the serotonin 1a receptor (5-HT1a, rs6295). Results showed the OPRM1 G-allele was associated with decreased CPM. A significant gene-to-gene interaction was found between the OPRM1 and the 5-HT1a gene. Reduced CPM scores were seen particularly in individuals with the OPRM1 G*/5-HT1a CC genotype, indicating that the 5-HT1a CC genotype seems to have an inhibiting effect on CPM if an individual has the OPRM1 G-genotype. Thus, regardless of pain phenotype, the OPRM1 G-allele independently as well as with an interaction with the 5-HT1a gene influenced pain modulation. FM patients had lower CPM than HC but no group differences were found regarding the genetic effects on CPM, indicating that the results reflect more general mechanisms influencing pain modulatory processes rather than underlying the dysfunction of CPM in FM. In conclusion, a genetic variant known to alter the expression of, and binding to, the my-opioid receptor reduced a subject’s ability to activate descending pain inhibition. Also, the results suggest a genetically inferred gene-to-gene interaction between the main opioid receptor and a serotonergic structure essential for 5-HT transmission to modulate pain inhibition. The results in this study highlight the importance of studying joint synergistic and antagonistic effects of neurotransmittor systems in regard to pain modulation.
Collapse
Affiliation(s)
- Jeanette Tour
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology, Blekinge Hospital, Karlskrona, Sweden
- * E-mail:
| | - Angelica Sandström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Diana Kadetoff
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Stockholm Spine Center, Löwenströmska Hospital, Upplands Väsby, Sweden
| | - Martin Schalling
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Eva Kosek
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
24
|
Xie X, Gu J, Zhuang D, Zhou Y, Chen X, Shen W, Li L, Liu Y, Xu W, Hong Q, Xu Z, Chen W, Zhou W, Liu H. Association between rs1799971 in the mu opioid receptor gene and methadone maintenance treatment response. J Clin Lab Anal 2022; 36:e24750. [PMID: 36305091 PMCID: PMC9701885 DOI: 10.1002/jcla.24750] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/28/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Genetic variations can affect individual response to methadone maintenance treatment (MMT) for heroin addiction. The A118G variant (rs1799971) in the mu opioid receptor gene (OPRM1) is a potential candidate single nucleotide polymorphism (SNP) for personalized MMT. This study determined whether rs1799971 is related to MMT response or dose. METHODS We recruited 286 MMT patients from a Han Chinese population. The rs1799971 genotype was determined via TaqMan genotyping assay. The genetic effect of this SNP on MMT response or dose was evaluated using logistic regression. A meta-analysis was performed to merge all available data to evaluate the role of rs1799971 in MMT using RevMan 5.3 software. RESULTS No statistical significance was observed in the association between the OPRM1 rs1799971 and MMT response or dose in our Chinese cohort. Meta-analysis indicated that the OPRM1 A118G variation was not significantly associated with MMT response or dose requirement. CONCLUSION The results suggest that rs1799971 in OPRM1 might not play a critical role alone in influencing MMT response or dose.
Collapse
Affiliation(s)
- Xiaohu Xie
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
| | - Jun Gu
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
| | - Dingding Zhuang
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
| | - Yun Zhou
- School of Medicine Ningbo University Ningbo China
| | - Xiaoyu Chen
- School of Medicine Ningbo University Ningbo China
| | - Wenwen Shen
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
| | - Longhui Li
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
| | - Yue Liu
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
| | - Wenjin Xu
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
| | - Qingxiao Hong
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
| | - Zemin Xu
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
| | - Weisheng Chen
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
| | - Wenhua Zhou
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
- School of Medicine Ningbo University Ningbo China
| | - Huifen Liu
- Key Laboratory of Addiction Research of Zhejiang Province Ningbo Kangning Hospital Ningbo China
- School of Medicine Ningbo University Ningbo China
| |
Collapse
|
25
|
Ye F, Du L, Huang W, Wang S. Shared Genetic Regulatory Networks Contribute to Neuropathic and Inflammatory Pain: Multi-Omics Systems Analysis. Biomolecules 2022; 12:1454. [PMID: 36291662 PMCID: PMC9599593 DOI: 10.3390/biom12101454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/27/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
The mechanisms of chronic pain are complex, and genetic factors play an essential role in the development of chronic pain. Neuropathic pain (NP) and inflammatory pain (IP) are two primary components of chronic pain. Previous studies have uncovered some common biological processes in NP and IP. However, the shared genetic mechanisms remained poorly studied. We utilized multi-omics systematic analyses to investigate the shared genetic mechanisms of NP and IP. First, by integrating several genome-wide association studies (GWASs) with multi-omics data, we revealed the significant overlap of the gene co-expression modules in NP and IP. Further, we uncovered the shared biological pathways, including the previously reported mitochondrial electron transport and ATP metabolism, and stressed the role of genetic factors in chronic pain with neurodegenerative diseases. Second, we identified 24 conservative key drivers (KDs) contributing to NP and IP, containing two well-established pain genes, IL1B and OPRM1, and some novel potential pain genes, such as C5AR1 and SERPINE1. The subnetwork of those KDs highlighted the processes involving the immune system. Finally, gene expression analysis of the KDs in mouse models underlined two of the KDs, SLC6A15 and KCNQ5, with unidirectional regulatory functions in NP and IP. Our study provides strong evidence to support the current understanding of the shared genetic regulatory networks underlying NP and IP and potentially benefit the future common therapeutic avenues for chronic pain.
Collapse
Affiliation(s)
- Fang Ye
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Li Du
- Vitalant Research Institute, San Francisco, CA 94118, USA
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA
| | - Wenqi Huang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Sheng Wang
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
26
|
Kaya-Akyüzlü D, Özkan-Kotiloğlu S, Bal C, Avcıoğlu G, Yalçın-Şahiner Ş, Şahiner İV. Sublingual buprenorphine/naloxone treatment is not affected by OPRM1 A118G and BDNF Va66Met polymorphisms, but alters the plasma beta-endorphin and BDNF levels in individuals with opioid use disorder. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 95:103979. [PMID: 36174909 DOI: 10.1016/j.etap.2022.103979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 06/16/2023]
Abstract
The study aimed to examine the genetic contribution to buprenorphine (BUP) treatment in individuals with opioid use disorder (OUD), with a specific focus on BDNF and OPRM1 genes. A total of 113 controls and 111 OUD patients receiving sublingual BUP/naloxone were enrolled. OPRM1 A118G and BDNF Val66Met polymorphisms were investigated by PCR-FRLP. Plasma BDNF and beta-endorphin levels were assessed by ELISA kits in both groups. Blood BUP levels were measured by LC-MS/MS and normalized with daily BUP dose (BUP/D). OPRM1 A118G and BDNF Val66Met polymorphisms didn't have an effect on plasma beta-endorphin and BDNF levels in OUD patients, respectively. Interestingly, OUD patients had significantly higher plasma BDNF and lower beta-endorphin levels compared to the controls (p < 0.001). A negative and significant correlation between plasma BUP/D and BDNF levels was found. Age onset of first use was associated with OPRM1 A118G polymorphism. The findings indicated that sublingual BUP/naloxone may increase plasma BDNF levels, but may decrease beta-endorphin levels in individuals with OUD. Plasma BDNF level seemed to be decreased in a BUP/D concentration-dependent manner.
Collapse
Affiliation(s)
| | - Selin Özkan-Kotiloğlu
- Kırşehir Ahi Evran University, Faculty of Science and Art, Department of Molecular Biology and Genetics, Kırşehir, Turkey
| | - Ceylan Bal
- Ankara Yıldırım Beyazıt University, Department of Medical Biochemistry, Ankara, Turkey
| | - Gamze Avcıoğlu
- Ankara Yıldırım Beyazıt University, Department of Medical Biochemistry, Ankara, Turkey
| | | | | |
Collapse
|
27
|
Fernández-de-las-Peñas C, Giordano R, Díaz-Gil G, Gil-Crujera A, Gómez-Sánchez SM, Ambite-Quesada S, Arendt-Nielsen L. Are Pain Polymorphisms Associated with the Risk and Phenotype of Post-COVID Pain in Previously Hospitalized COVID-19 Survivors? Genes (Basel) 2022; 13:genes13081336. [PMID: 35893072 PMCID: PMC9394327 DOI: 10.3390/genes13081336] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 02/01/2023] Open
Abstract
Objective: To investigate the association of different, selected pain polymorphisms with the presence of de novo long-COVID pain symptoms and to analyze the association between these polymorphisms with clinical, sensory-related, cognitive and psychological variables in COVID-19 survivors. Methods: Two hundred and ninety-three (n = 293, 49.5% female, mean age: 55.6 ± 12.9 years) previously hospitalized COVID-19 survivors participated. Three genotypes of the following single nucleotide polymorphisms (SNPs) were obtained from non-stimulated saliva: OPRM1 (rs1799971), COMT (rs4680), BDNF (rs6265), and HTR1B (rs6296) by polymerase chain reactions in all participants. Further, clinical (intensity/duration of pain), sensory-related (sensitization-associated symptoms, neuropathic pain features), psychological (anxiety or depressive levels, sleep quality), and cognitive (catastrophizing, kinesiophobia) variables were collected in those COVID-19 survivors suffering from post-COVID pain. Analyses were carried out to associate clinical features with genotype. Results: Participants were assessed 17.8 ± 5.2 months after hospitalization. One hundred and seventeen (39.9%) experienced post-COVID pain (particularly of musculoskeletal origin). The distributions of the genotype variants of any SNP were not significantly different between COVID-19 survivors with and without long-term post-COVID pain (all, p > 0.178). No differences in sensitization-associated symptoms, neuropathic pain features, catastrophizing, kinesiophobia levels, anxiety and depressive levels or sleep quality according to the genotype variant in any SNPs were found. No effect of gender was identified. Conclusion: The four SNPs generally associated with pain did not appear to predispose to the development of de novo long-COVID pain symptoms in previously hospitalized COVID-19 survivors. The SNPs were not involved in the phenotypic features of post-COVID pain either.
Collapse
Affiliation(s)
- César Fernández-de-las-Peñas
- Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain;
- Center for Neuroplasticity and Pain (CNAP), SMI, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, DK-9220 Aalborg, Denmark; (R.G.); (L.A.-N.)
- Correspondence:
| | - Rocco Giordano
- Center for Neuroplasticity and Pain (CNAP), SMI, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, DK-9220 Aalborg, Denmark; (R.G.); (L.A.-N.)
| | - Gema Díaz-Gil
- Research Group GAMDES, Department of Basic Health Sciences, Universidad Rey Juan Carlos (URJC), 28922 Madrid, Spain; (G.D.-G.); (A.G.-C.); (S.M.G.-S.)
| | - Antonio Gil-Crujera
- Research Group GAMDES, Department of Basic Health Sciences, Universidad Rey Juan Carlos (URJC), 28922 Madrid, Spain; (G.D.-G.); (A.G.-C.); (S.M.G.-S.)
| | - Stella M. Gómez-Sánchez
- Research Group GAMDES, Department of Basic Health Sciences, Universidad Rey Juan Carlos (URJC), 28922 Madrid, Spain; (G.D.-G.); (A.G.-C.); (S.M.G.-S.)
| | - Silvia Ambite-Quesada
- Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain;
| | - Lars Arendt-Nielsen
- Center for Neuroplasticity and Pain (CNAP), SMI, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, DK-9220 Aalborg, Denmark; (R.G.); (L.A.-N.)
- Department of Medical Gastroenterology, Mech-Sense, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| |
Collapse
|
28
|
Can the Type of Subjective Response to First Opioid Exposure Predict the Risk of Opioid Use Disorder? A Scoping Review. Brain Res Bull 2022; 188:67-76. [PMID: 35901984 DOI: 10.1016/j.brainresbull.2022.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/07/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Effective identification of individuals at increased risk for developing opioid use disorder (OUD) could reduce the overdose fatalities and mitigate the harm of the opioid epidemic. Early evidence has linked certain subjective experiences during the first substance exposure to subsequent substance misuse. This is consistent with anecdotal evidence that "first response" to opioids may distinguish those who later develop OUD from those who do not, further suggesting individual differences in neural activity and responses to specific substances. OBJECTIVE The objective of this scoping review was to evaluate the evidence on the relationship between the subjective first response to opioids and the risk of developing OUD. METHODS Ovid MEDLINE search (through 1/29/2021), followed by the search for articles published by the first/senior authors (PubMed) and references citing (Web of Science Citing Reference) identified eligible publications, was focused on studies involving humans, published in English, and describing the initial subjective response to opioid exposure and its association with OUD development or its risk. Systematic data extraction was completed for each eligible study. RESULTS 3,364 title/abstracts and 48 full-text articles were reviewed. Four articles, describing six studies (526 adult participants) were eligible and included. These studies were methodologically heterogeneous, including 2 randomized controlled trials, and 1 case-control, and 3 cross-sectional studies, with small sample sizes, precluding data pooling and meta-analysis. Results suggested that individuals with OUD or at elevated risk for OUD (determined by the validated OUD risk assessment surveys) were more likely to report their "first opioid response" as euphoric compared to their counterparts. CONCLUSIONS Limited evidence suggests that subjectively-reported euphoric response to first opioid exposure could predict the OUD risk. Prospective studies are needed to evaluate the utility of assessing first response (e.g., via brief point-of-care screening surveys) for informing clinical decisions when prescribing opioids.
Collapse
|
29
|
Yee L, Capule FR, Makmor-Bakry M. Genetic polymorphisms of OPRM1 on the efficacy and safety of anesthetic and analgesic agents: a systematic review. Pharmacogenomics 2022; 23:609-617. [PMID: 35735174 DOI: 10.2217/pgs-2022-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This systematic review aimed to outline the outcome of OPRMI (A118G) variants on the effects of anesthetic and analgesic agents used in various procedures. Materials & methods: Literature was obtained from reliable, established databases and reference tracking. Efficacy and side/adverse effects of anesthetic and analgesic drugs intraoperatively or within 48 h postsurgery were the key outcome measures for all populations. Animal studies were excluded. Results: Twenty-nine studies were chosen for inclusion. In association with the efficacy and safety of anesthetic and analgesic agents, gene polymorphism in OPRM1 displayed a strong correlation in reduced analgesic effect and protection against adverse reactions. Conclusion: This systematic review summarized the correlation between genetic polymorphism in the OPRM1 gene and anesthetic/analgesic effects.
Collapse
Affiliation(s)
- Liew Yee
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia
| | - Francis R Capule
- College of Pharmacy. University of the Philippines Manila, Manila, 1000, Philippines
| | - Mohd Makmor-Bakry
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia
| |
Collapse
|
30
|
Ayuso P, Macías Y, Gómez-Tabales J, García-Martín E, Agúndez JAG. Molecular monitoring of patient response to painkiller drugs. Expert Rev Mol Diagn 2022; 22:545-558. [PMID: 35733288 DOI: 10.1080/14737159.2022.2093638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Non-steroidal anti-inflammatory drugs and opioids are widely prescribed for the treatment of mild to severe pain. Wide interindividual variability regarding the analgesic efficacy and adverse reactions to these drugs (ADRs) exist, although the mechanisms responsible for these ADRs are not well understood. AREAS COVERED We provide an overview of the clinical impact of variants in genes related to the pharmacokinetics and pharmacodynamics of painkillers, as well as those associated with the susceptibility to ADRs. Also, we discuss the current pharmacogenetic-guided treatment recommendations for the therapeutic use of non-steroidal anti-inflammatory drugs and opioids. EXPERT OPINION In the light of the data analyzed, common variants in genes involved in pharmacokinetics and pharmacodynamics processes may partially explain the lack of response to painkiller treatment and the occurrence of adverse drug reactions. The implementation of high-throughput sequencing technologies may help to unveil the role of rare variants as considerable contributors to explaining the interindividual variability in drug response. Furthermore, a consensus between the diverse pharmacogenetic guidelines is necessary to extend the implementation of pharmacogenetic-guided prescription in daily clinical practice. Additionally, the physiologically-based pharmacokinetics and pharmacodynamics modeling techniques may contribute to the improvement of these guidelines and facilitate clinicians drug dose adjustment.
Collapse
Affiliation(s)
- Pedro Ayuso
- University Institute of Molecular Pathology Biomarkers, UEx. ARADyAL, Instituto de Salud Carlos III, Cáceres, Spain
| | - Yolanda Macías
- University Institute of Molecular Pathology Biomarkers, UEx. ARADyAL, Instituto de Salud Carlos III, Cáceres, Spain
| | - Javier Gómez-Tabales
- University Institute of Molecular Pathology Biomarkers, UEx. ARADyAL, Instituto de Salud Carlos III, Cáceres, Spain
| | - Elena García-Martín
- University Institute of Molecular Pathology Biomarkers, UEx. ARADyAL, Instituto de Salud Carlos III, Cáceres, Spain
| | - José A G Agúndez
- University Institute of Molecular Pathology Biomarkers, UEx. ARADyAL, Instituto de Salud Carlos III, Cáceres, Spain
| |
Collapse
|
31
|
Firfirey F, September AV, Shamley D. ABCB1 and OPRM1 single-nucleotide polymorphisms collectively modulate chronic shoulder pain and dysfunction in South African breast cancer survivors. Pharmacogenomics 2022; 23:513-530. [PMID: 35727214 DOI: 10.2217/pgs-2022-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: Chronic shoulder pain/disability is a well-recognized side effect of treatment for breast cancer, with ∼40% of patients experiencing this, despite receiving pain management. To manage acute and chronic pain, several opioids are commonly prescribed. Pharmacogenomics have implicated genes within the opioid signaling pathway, including ABCB1 and OPRM1, to contribute to an individual's variable response to opioids. Aim: To evaluate ABCB1 (rs1045642 G>A, rs1128503 G>A) and OPRM1 (rs1799971 A>G, rs540825 T>A) single-nucleotide polymorphisms (SNPs) in chronic shoulder pain/disability in BCS. Materials & methods: TaqManTM assays were used to genotype ABCB1 and OPRM1 SNPs within the BCS (N = 252) cohort. The Shoulder Pain and Disability Index was used to evaluate pain and disability features associated with shoulder pathologies. Participants end scores for each feature (pain, disability and combined [pain and disability]) were categorized into no-low (>30%) and moderate-high (≥30%) scores. Statistical analysis was applied, and significance was accepted at p < 0.05. Results: Of participants, 27.0, 19.0 and 22.0% reported moderate-high pain, disability and combined (pain and disability) scores, respectively. ABCB1:rs1045642-(A/A) genotype was significantly associated with disability (p = 0.028: no-low [14.9%] vs mod-high [4.3%]) and combined (pain and disability) (p = 0.011: no-low [15.9%] vs mod-high [5.7%]). The ABCB1:rs1045642-(A) allele was significantly associated with disability (p = 0.015: no-low [37.9%] vs mod-high [23.9%]) and combined (pain and disability) (p = 0.003: no-low [38.5%] vs mod-high [23.6%]). The inferred ABCB1 (rs1045642 G>A - rs1128503 G>A): A-G (p = 0.029; odds ratio [OR]: 0.0; 95% CI: 0.0-0.0) and the OPRM1 (rs1799971 A>G - rs540825 T>A): G-T (p = 0.019; OR: 0.33; 95% CI: 0.14-0.75) haplotypes were associated with disability and pain, respectively. Gene-gene interactions showed the ABCB1 (rs1045642 G>A) - OPRM1 (rs540825 T>A) combinations, (A-T) (p = 0.019; OR: 0.62; 95% CI: 0.33-1.16) and (G-A) (p = 0.021; OR: 1.57; 95% CI: 0.30-3.10) were associated with disability. Conclusion: The study implicated ABCB1 with shoulder pain and disability; and haplotype analyses identified specific genetic intervals within ABCB1 and OPRM1 to associate with chronic shoulder pain and disability. Evidence suggests that potentially gene-gene interactions between ABCB1 and OPRM1 contribute to chronic shoulder pain and disability experienced in this SA cohort.
Collapse
Affiliation(s)
- Firzana Firfirey
- Department of Human Biology, Division of Physiological Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, Western Cape, 7701, South Africa
| | - Alison V September
- Department of Human Biology, Division of Physiological Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, Western Cape, 7701, South Africa.,Department of Human Biology, Health through Physical Activity, Lifestyle & Sport Research Centre (HPALS), Faculty of Health Sciences, University of Cape Town, Cape Town, Western Cape, 7701, South Africa.,Department of Human Biology, International Federation of Sports Medicine (FIMS), Collaborative Centre of Sports Medicine, University of Cape Town, Cape Town, Western Cape, 7701, South Africa
| | - Delva Shamley
- Department of Human Biology, Division of Clinical Anatomy & Biological Anthropology, Anatomy Building, Medical School, University of Cape Town, Cape Town, Western Cape, 7701, South Africa
| |
Collapse
|
32
|
Cahill S, Chandola T, Hager R. Genetic Variants Associated With Resilience in Human and Animal Studies. Front Psychiatry 2022; 13:840120. [PMID: 35669264 PMCID: PMC9163442 DOI: 10.3389/fpsyt.2022.840120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
Resilience is broadly defined as the ability to maintain or regain functioning in the face of adversity and is influenced by both environmental and genetic factors. The identification of specific genetic factors and their biological pathways underpinning resilient functioning can help in the identification of common key factors, but heterogeneities in the operationalisation of resilience have hampered advances. We conducted a systematic review of genetic variants associated with resilience to enable the identification of general resilience mechanisms. We adopted broad inclusion criteria for the definition of resilience to capture both human and animal model studies, which use a wide range of resilience definitions and measure very different outcomes. Analyzing 158 studies, we found 71 candidate genes associated with resilience. OPRM1 (Opioid receptor mu 1), NPY (neuropeptide Y), CACNA1C (calcium voltage-gated channel subunit alpha1 C), DCC (deleted in colorectal carcinoma), and FKBP5 (FKBP prolyl isomerase 5) had both animal and human variants associated with resilience, supporting the idea of shared biological pathways. Further, for OPRM1, OXTR (oxytocin receptor), CRHR1 (corticotropin-releasing hormone receptor 1), COMT (catechol-O-methyltransferase), BDNF (brain-derived neurotrophic factor), APOE (apolipoprotein E), and SLC6A4 (solute carrier family 6 member 4), the same allele was associated with resilience across divergent resilience definitions, which suggests these genes may therefore provide a starting point for further research examining commonality in resilience pathways.
Collapse
Affiliation(s)
- Stephanie Cahill
- Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Faculty of Humanities, Cathie Marsh Institute for Social Research, The University of Manchester, Manchester, United Kingdom
| | - Tarani Chandola
- Faculty of Humanities, Cathie Marsh Institute for Social Research, The University of Manchester, Manchester, United Kingdom
- Methods Hub, Department of Sociology, Faculty of Social Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Reinmar Hager
- Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
33
|
Gondré-Lewis MC, Elman I, Alim T, Chapman E, Settles-Reaves B, Galvao C, Gold MS, Baron D, Kazmi S, Gardner E, Gupta A, Dennen C, Blum K. Frequency of the Dopamine Receptor D3 (rs6280) vs. Opioid Receptor µ1 (rs1799971) Polymorphic Risk Alleles in Patients with Opioid Use Disorder: A Preponderance of Dopaminergic Mechanisms? Biomedicines 2022; 10:biomedicines10040870. [PMID: 35453620 PMCID: PMC9027142 DOI: 10.3390/biomedicines10040870] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/21/2022] [Accepted: 03/26/2022] [Indexed: 02/05/2023] Open
Abstract
While opioids are a powerful class of drugs that inhibit transmission of pain signals, their use is tarnished by the current epidemic of opioid use disorder (OUD) and overdose deaths. Notwithstanding published reports, there remain gaps in our knowledge of opioid receptor mechanisms and their role in opioid seeking behavior. Thus, novel insights into molecular, neurogenetic and neuropharmacological bases of OUD are needed. We propose that an addictive endophenotype may not be entirely specific to the drug of choice but rather may be generalizable to altered brain reward circuits impacting net mesocorticolimbic dopamine release. We suggest that genetic or epigenetic alterations across dopaminergic reward systems lead to uncontrollable self-administration of opioids and other drugs. For instance, diminished availability via knockout of dopamine D3 receptor (DRD3) increases vulnerability to opioids. Building upon this concept via the use of a sophisticated polymorphic risk analysis in a human cohort of chronic opioid users, we found evidence for a higher frequency of polymorphic DRD3 risk allele (rs6280) than opioid receptor µ1 (rs1799971). In conclusion, while opioidergic mechanisms are involved in OUD, dopamine-related receptors may have primary influence on opioid-seeking behavior in African Americans. These findings suggest OUD-targeted novel and improved neuropharmacological therapies may require focus on DRD3-mediated regulation of dopaminergic homeostasis.
Collapse
Affiliation(s)
- Marjorie C. Gondré-Lewis
- Neuropsychopharmacology Laboratory, Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA (C.G.)
- Correspondence: (M.C.G.-L.); (K.B.)
| | - Igor Elman
- Department of Psychiatry, Cambridge Health Alliance/Harvard Medical School, Cambridge, MA 02139, USA or
| | - Tanya Alim
- Department of Psychiatry and Behavioral Sciences, Howard University College of Medicine, Washington, DC 20059, USA; (T.A.); (E.C.)
| | - Edwin Chapman
- Department of Psychiatry and Behavioral Sciences, Howard University College of Medicine, Washington, DC 20059, USA; (T.A.); (E.C.)
| | - Beverlyn Settles-Reaves
- Neuropsychopharmacology Laboratory, Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA (C.G.)
| | - Carine Galvao
- Neuropsychopharmacology Laboratory, Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA (C.G.)
| | - Mark S. Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - David Baron
- Graduate College, Western University Health Sciences, Pomona, CA 91766, USA;
| | - Shan Kazmi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA or
| | - Eliot Gardner
- Neuropsychopharmacology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA;
| | - Ashim Gupta
- Future Biologics, Lawrenceville, GA 30043, USA;
| | - Catherine Dennen
- The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX 78701, USA;
| | - Kenneth Blum
- Graduate College, Western University Health Sciences, Pomona, CA 91766, USA;
- The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX 78701, USA;
- Department of Clinical Psychology and Addiction, Institute of Psychology, Faculty of Education and Psychology, ELTE Eötvös Loránd University, Egyetem tér 1-3, 1053 Budapest, Hungary
- Department of Psychiatry, School of Medicine, University of Vermont, Burlington, VT 05405, USA
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur 721172, West Bengal, India
- Department of Psychiatry, Wright State University Boonshoft School of Medicine and Dayton VA Medical Centre, Dayton, OH 45324, USA
- Correspondence: (M.C.G.-L.); (K.B.)
| |
Collapse
|
34
|
Baker Rogers J, Higa GM. Spoken and Unspoken Matters Regarding the Use of Opioids in Cancer. J Pain Res 2022; 15:909-924. [PMID: 35411188 PMCID: PMC8994621 DOI: 10.2147/jpr.s349107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/24/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Janna Baker Rogers
- Sections of Geriatrics, Palliative Medicine and Hospice, Department of Medicine, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Gerald M Higa
- Departments of Clinical Pharmacy and Medicine, Schools of Pharmacy and Medicine, West Virginia University, Morgantown, WV, USA
- Correspondence: Gerald M Higa, Departments of Clinical Pharmacy and Medicine, Schools of Pharmacy and Medicine, West Virginia University, Morgantown, WV, USA, 26506, Email
| |
Collapse
|
35
|
Hriatpuii V, Sema HP, Vankhuma C, Iyer M, Subramaniam MD, Rao KRSS, Vellingiri B, Kumar NS. Association of OPRM1 with addiction: a review on drug, alcohol and smoking addiction in worldwide population. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00249-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Drugs are chemicals which can disrupt the nerve cell functions of the brain. The present study aims to investigate the addiction related gene (OPRM1) in three types of addiction—drugs, alcohol and smoking. Pathway for the addiction was ascertained through KEGG database, and the hotspot mutations for various populations were identified from Gnomad-exomes database. In silico analyses like SIFT, Polyphen, Hope, I-mutant and mutation taster were performed to understand the amino acid substitution, protein function, stability and pathogenicity of the variants.
Main body
Addiction-related variants were found in exons 1, 2 and 3, while the exon 4 did not exhibit any addiction related variation. Among all the variants from this gene, rs1799971 (A118G) polymorphism was the most commonly studied variation for addiction in different populations worldwide. Population-wise allele and genotype frequencies, demographic and epidemiological studies have also been performed from different populations, and the possible association of these variants with addiction was evaluated.
Conclusion
Our findings suggest that OPRM1 polymorphism impact as pharmacogenetic predictor of response to naltrexone and can also address the genetic predisposition related to addiction in human beings.
Collapse
|
36
|
Wood EK, Baron Z, Schwandt ML, Lindell SG, Barr CS, Suomi SJ, Higley JD. Variation in the Mu-Opioid Receptor (OPRM1) and Offspring Sex Are Associated With Maternal Behavior in Rhesus Macaques (Macaca mulatta). Front Behav Neurosci 2022; 16:721958. [PMID: 35368303 PMCID: PMC8964435 DOI: 10.3389/fnbeh.2022.721958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 01/06/2022] [Indexed: 12/14/2022] Open
Abstract
A μ-opioid receptor (OPRM1) single-nucleotide-polymorphism, found in both humans and rhesus macaques mediates the mother-infant attachment bond. Because mothers treat their sons and daughters differently, it is somewhat surprising that the role of infant sex has not been assessed in the context of a maternal-OPRM1-genotype-by-infant-sex interaction. The present study investigates the effect of maternal-OPRM1-genotype and infant sex on mother-infant behaviors. Over the first 6 months of offspring life, mother-infant behavioral data assessing attachment quality was collected twice weekly from a large number of rhesus monkey mother-infant pairs (N = 161 dyads; n = 64 female infants, n = 97 male infants). Mothers were genotyped for OPRM1 variation. Factor analysis of the observed behaviors showed two factors: Attachment (maternal-infant cradling, rejections, and infant approaches and leaves), and Maternal Restraints (mother restrains infant, preventing exploration). Further analyses showed a two-way, maternal-genotype-by-infant-sex interaction for both factors. For Attachment, mothers with the CC genotype cradled and restrained (Maternal Restraints) their female infants more and rejected them less, when compared to female infants of CG mothers. Perhaps as a consequence, female infants of CC genotype mothers approached and left their mothers less often, when compared to female infants of CG mothers, likely an indication that female infants from mothers with CG genotype play a greater role in maintaining the mother-infant bond than do female infants from CC genotype mothers. This finding may also indicate a more secure attachment in infants from CC genotype mothers. Unlike female infants, on average, the mother-infant relationship of dyads with a male infant was largely undifferentiated by maternal genotype. These findings suggest that, in contrast to female infants from CG mothers, CC mothers and their female infants appear to have a closer mother-infant relationship which may portend close life-long bonds, as mothers and female offspring remain together throughout life. Male offspring appear to have a more aloof mother-infant bond regardless of OPRM1-genotype. The results of this study indicate that maternal-OPRM1 variation mediates mother-infant attachment behaviors for female infants and has less effect for male infants. This suggests that offspring sex should be included in studies investigating the effect of maternal-OPRM1 genotype on the mother-infant attachment relationship.
Collapse
Affiliation(s)
- Elizabeth K. Wood
- Department of Psychology, Brigham Young University, Provo, UT, United States
| | - Zachary Baron
- Department of Neuroscience, Brigham Young University, Provo, UT, United States
| | - Melanie L. Schwandt
- National Institute on Alcohol Abuse and Alcoholism (NIAAA), Bethesda, MD, United States
| | - Stephen G. Lindell
- National Institute on Alcohol Abuse and Alcoholism (NIAAA), Bethesda, MD, United States
| | - Christina S. Barr
- National Institute on Alcohol Abuse and Alcoholism (NIAAA), Bethesda, MD, United States
| | - Stephen J. Suomi
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - J. Dee Higley
- Department of Psychology, Brigham Young University, Provo, UT, United States
- *Correspondence: J. Dee Higley,
| |
Collapse
|
37
|
Tabata Y, Matsuo Y, Fujii Y, Ohta A, Hirota K. Rapid detection of single nucleotide polymorphisms using the MinION nanopore sequencer: a feasibility study for perioperative precision medicine. JA Clin Rep 2022; 8:17. [PMID: 35244794 PMCID: PMC8897523 DOI: 10.1186/s40981-022-00506-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 11/10/2022] Open
Abstract
Introduction Precision medicine is a phrase used to describe personalized medical care tailored to specific patients based on their clinical presentation and genetic makeup. However, despite the fact that several single nucleotide polymorphisms (SNPs) have been reported to be associated with increased susceptibility to particular anesthetic agents and the occurrence of perioperative complications, genomic profiling and thus precision medicine has not been widely applied in perioperative management. Methods We validated six SNP loci known to affect perioperative outcomes in Japanese patients using genomic DNA from saliva specimens and nanopore sequencing of each SNP loci to facilitate allele frequency calculations and then compared the nanopore results to those produced using the conventional dideoxy sequencing method. Results Nanopore sequencing reads clustered into the expected genotypes in both homozygous and heterozygous cases. In addition, the nanopore sequencing results were consistent with those obtained using conventional dideoxy sequencing and the workflow provided reliable allele frequency estimation, with a total analysis time of less than 4 h. Conclusion Thus, our results suggest that nanopore sequencing is a promising and versatile tool for SNP genotyping, allowing for rapid and feasible risk prediction of perioperative outcomes. Supplementary Information The online version contains supplementary material available at 10.1186/s40981-022-00506-7.
Collapse
Affiliation(s)
- Yoshiteru Tabata
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| | - Yoshiyuki Matsuo
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan.
| | - Yosuke Fujii
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan.,Department of Anesthesia, Otsu City Hospital, Otsu, Shiga, Japan
| | - Atsufumi Ohta
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| | - Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
38
|
Custodio L, Malone S, Bardo MT, Turner JR. Nicotine and opioid co-dependence: Findings from bench research to clinical trials. Neurosci Biobehav Rev 2022; 134:104507. [PMID: 34968525 PMCID: PMC10986295 DOI: 10.1016/j.neubiorev.2021.12.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 10/19/2022]
Abstract
Concomitant use of tobacco and opioids represents a growing public health concern. In fact, the mortality rate due to smoking-related illness approaches 50% among SUD patients. Cumulative evidence demonstrates that the vulnerability to drugs of abuse is influenced by behavioral, environmental, and genetic factors. This review explores the contribution of genetics and neural mechanisms influencing nicotine and opioid reward, respiration, and antinociception, emphasizing the interaction of cholinergic and opioid receptor systems. Despite the substantial evidence demonstrating nicotine-opioid interactions within the brain and on behavior, the currently available pharmacotherapies targeting these systems have shown limited efficacy for smoking cessation on opioid-maintained smokers. Thus, further studies designed to identify novel targets modulating both nicotinic and opioid receptor systems may lead to more efficacious approaches for co-morbid nicotine dependence and opioid use disorder.
Collapse
Affiliation(s)
- Lilian Custodio
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Samantha Malone
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | - Michael T Bardo
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | - Jill R Turner
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
39
|
Epigenetic moderators of naltrexone efficacy in reducing heavy drinking in Alcohol Use Disorder: a randomized trial. THE PHARMACOGENOMICS JOURNAL 2022; 22:1-8. [PMID: 34381173 PMCID: PMC8799481 DOI: 10.1038/s41397-021-00250-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023]
Abstract
Polymorphisms in genes associated with opioid signaling and dopamine reuptake and inactivation may moderate naltrexone efficacy in Alcohol Use Disorder (AUD), but the effects of epigenetic modification of these genes on naltrexone response are largely unexplored. This study tested interactions between methylation in the μ-opioid receptor (OPRM1), dopamine transporter (SLC6A3), and catechol-O-methyltransferase (COMT) genes as predictors of naltrexone effects on heavy drinking in a 16-week randomized, placebo-controlled trial among 145 treatment-seeking AUD patients. OPRM1 methylation interacted with both SLC6A3 and COMT methylation to moderate naltrexone efficacy, such that naltrexone-treated individuals with lower methylation of the OPRM1 promoter and the SLC6A3 promoter (p = 0.006), COMT promoter (p = 0.005), or SLC6A3 3' untranslated region (p = 0.004), relative to placebo and to those with higher OPRM1 and SLC6A3 or COMT methylation, had significantly fewer heavy drinking days. Epigenetic modification of opioid- and dopamine-related genes may represent a novel pharmacoepigenetic predictor of naltrexone efficacy in AUD.
Collapse
|
40
|
Lans J, Westenberg RF, Gottlieb RE, Valerio IL, Chen NC, Eberlin KR. Long-Term Opioid Use Following Surgery for Symptomatic Neuroma. J Reconstr Microsurg 2022; 38:137-143. [PMID: 35100646 DOI: 10.1055/s-0041-1731640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Identifying patients at risk for prolonged opioid use following surgery for symptomatic neuroma would be beneficial for perioperative management. The aim of this study is to identify the factors associated with postoperative opioid use of >4 weeks in patients undergoing neuroma surgery. METHODS After retrospective identification, 77 patients who underwent surgery for symptomatic neuroma of the upper or lower extremity were enrolled. Patients completed the Patient-Reported Outcomes Measurement Information System (PROMIS) depression, Numeric Rating Scale (NRS) pain score, and a custom medication questionnaire at a median of 9.7 years (range: 2.5-16.8 years) following surgery. Neuroma excision followed by nerve implantation (n = 39, 51%), nerve reconstruction/repair (n = 18, 23%), and excision alone (n = 16, 21%) were the most common surgical treatments. RESULTS Overall, 27% (n = 21) of patients reported opioid use of more than 4 weeks postoperatively. Twenty-three patients (30%) reported preoperative opioid use of which 11 (48%) did not report opioid use for >4 weeks, postoperatively. In multivariable logistic regression, preoperative opioid use was independently associated with opioid use of >4 weeks, postoperatively (odds ratio [OR] = 4.4, 95% confidence interval [CI]: 1.36-14.3, p = 0.013). CONCLUSION Neuroma surgery reduces opioid use in many patients but patients who are taking opioids preoperatively are at risk for longer opioid use. Almost one-third of patients reported opioid use longer than 4 weeks, postoperatively.
Collapse
Affiliation(s)
- Jonathan Lans
- Department of Orthopaedic Surgery, Hand and Upper Extremity Service, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Ritsaart F Westenberg
- Department of Orthopaedic Surgery, Hand and Upper Extremity Service, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Rachel E Gottlieb
- Department of Orthopaedic Surgery, Hand and Upper Extremity Service, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Ian L Valerio
- Division of Plastic Surgery, Hand Surgery, and Peripheral Nerve Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Neal C Chen
- Department of Orthopaedic Surgery, Hand and Upper Extremity Service, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Kyle R Eberlin
- Division of Plastic Surgery, Hand Surgery, and Peripheral Nerve Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
41
|
Bourgault Z, Matheson J, Mann RE, Brands B, Wickens CM, Tiwari AK, Zai CC, Kennedy J, Le Foll B. Mu opioid receptor gene variant modulates subjective response to smoked cannabis. Am J Transl Res 2022; 14:623-632. [PMID: 35173880 PMCID: PMC8829626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
The mu-opioid receptor (MOR) mediates the rewarding properties of many psychoactive drugs and is an important target in the treatment of addictions. Functional interactions between the opioid and endocannabinoid systems are established and have been hypothesized to contribute to the effects of cannabis. We investigated associations between three single nucleotide polymorphisms in the MOR gene OPRM1 (rs1799971, rs2281617, and rs510769) and subjective responses to smoked cannabis. Fifty-two regular cannabis users (1-4 days/week) were given a cannabis cigarette (12.5% THC) and rated their subjective responses on visual analog scales at baseline and at multiple time points after smoking. Blood samples were collected for THC quantification. There was a significant impact of the intronic variant rs510769 on subjective cannabis effects and THC blood levels. The influence of this gene variant may thus be mediated by pharmacodynamics and/or pharmacokinetic factors. We provide novel evidence that variability in OPRM1 contributes to individual responses to cannabis and may affect risk of cannabis use disorder. Our findings add to the growing body of literature on the genetic basis of individual responses to cannabis and may have implications for targeting the endogenous opioid system in the treatment of cannabis use disorder.
Collapse
Affiliation(s)
- Zoe Bourgault
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, University of Toronto33 Ursula Franklin Street, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of TorontoToronto, ON, Canada
| | - Justin Matheson
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, University of Toronto33 Ursula Franklin Street, Toronto, ON, Canada
| | - Robert E Mann
- Institute for Mental Health Policy Research, Centre for Addiction and Mental HealthToronto, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental HealthToronto, Ontario, Canada
- Dalla Lana School of Public Health, University of TorontoToronto, Ontario, Canada
| | - Bruna Brands
- Department of Pharmacology and Toxicology, University of TorontoToronto, ON, Canada
- Institute for Mental Health Policy Research, Centre for Addiction and Mental HealthToronto, Canada
- Controlled Substances and Cannabis Directorate, Health CanadaOttawa, Canada
| | - Christine M Wickens
- Department of Pharmacology and Toxicology, University of TorontoToronto, ON, Canada
- Institute for Mental Health Policy Research, Centre for Addiction and Mental HealthToronto, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental HealthToronto, Ontario, Canada
- Dalla Lana School of Public Health, University of TorontoToronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, University of TorontoToronto, Ontario, Canada
| | - Arun K Tiwari
- Neurogenetics Section, Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research InstituteCAMH, Toronto, ON, Canada
- Department of Psychiatry, University of TorontoToronto, ON, Canada
| | - Clement C Zai
- Neurogenetics Section, Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research InstituteCAMH, Toronto, ON, Canada
- Department of Psychiatry, University of TorontoToronto, ON, Canada
- Institute of Medical Science, University of TorontoToronto, ON, Canada
- Laboratory Medicine and Pathobiology, University of TorontoON, Canada
- T.H. Chan School of Public Health, Harvard UniversityBoston, MA, USA
- Broad InstituteCambridge, MA, USA
| | - James Kennedy
- Neurogenetics Section, Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research InstituteCAMH, Toronto, ON, Canada
- Department of Psychiatry, University of TorontoToronto, ON, Canada
- Institute of Medical Science, University of TorontoToronto, ON, Canada
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, University of Toronto33 Ursula Franklin Street, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of TorontoToronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental HealthToronto, Ontario, Canada
- Department of Psychiatry, University of TorontoToronto, ON, Canada
- Institute of Medical Science, University of TorontoToronto, ON, Canada
- Acute Care Program, Centre for Addiction and Mental HealthToronto, ON, Canada
- Department of Family and Community Medicine, University of TorontoToronto, ON, Canada
| |
Collapse
|
42
|
Association of the D-amino acid oxidase gene with methadone dose in heroin dependent patients under methadone maintenance treatment. J Hum Genet 2022; 67:273-278. [PMID: 34983973 DOI: 10.1038/s10038-021-01008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 11/08/2022]
Abstract
Methadone is a synthetic opioid used for the maintenance treatment (MMT) of heroin dependence. It primarily binds to the μ-opioid receptor (MOR; with its gene, namely OPRM1). Methadone is also an N-methyl-D-aspartate (NMDA) receptor antagonist. The role of NMDA receptor in the regulatory mechanisms of methadone dosage in heroin dependent patients is so far not clear. D-amino acid oxidase (DAO) is an important enzyme that indirectly activates the NMDA receptor through its effect on the D-serine level. To test the hypothesis that genetic polymorphisms in the DAO gene are associated with methadone treatment dose and responses, we selected four single nucleotide polymorphisms (SNPs) in DAO from the literature reports of the Taiwanese population. SNPs were genotyped in 344 MMT patients. In this study, we identified a functional SNP rs55944529 in the DAO gene that reveals a modest but significant association with the methadone dosage in the recessive model of analysis (P = 0.003) and plasma concentrations (P = 0.003) in MMT patients. However, it did not show association with plasma methadone concentration in multiple linear regression analysis. It is also associated with the methadone adverse reactions of dry mouth (P = 0.002), difficulty with urination (P = 0.0003) in the dominant model, and the withdrawal symptoms of yawning (P = 0.005) and gooseflesh skin (P = 0.004) in the recessive model. Our results suggest a role of the indirect regulatory mechanisms of the NMDA reporter, possibly via the DAO genetic variants, in the methadone dose and some adverse reactions in MMT patients.
Collapse
|
43
|
Stoecker WV. Part I: Missouri's Fentanyl Poisonings Rise to Record Levels. MISSOURI MEDICINE 2022; 119:489-493. [PMID: 36588654 PMCID: PMC9762215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Missourians are dying of fentanyl poisoning at an unprecedented rate. We identified growth areas in Missouri for fatal fentanyl encounters in rural and western counties. Though the deaths occur for a multitude of reasons, a growing trend adds to the surge in fentanyl fatalities: poisonings from counterfeit pills. The tablets are often labeled with brand names for alprazolam or oxycodone, but may contain only fentanyl at a dangerous level. Teenagers find counterfeit pills all too easily via social media. Believing they have found an easy way to obtain a quick high or relief of minor pain and anxiety, they take the pill alone in their bedroom, with no possibility of reversing a fatal fentanyl dose. There is a wide range of respiratory depression from illicit drugs containing fentanyl. We reviewed the physiologic respiratory response to drugs containing fentanyl that varies with genetics and the unpredictable amount of fentanyl contained in illicit drugs.
Collapse
|
44
|
Brown TG, Xu J, Hurd YL, Pan YX. Dysregulated expression of the alternatively spliced variant mRNAs of the mu opioid receptor gene, OPRM1, in the medial prefrontal cortex of male human heroin abusers and heroin self-administering male rats. J Neurosci Res 2022; 100:35-47. [PMID: 32506472 PMCID: PMC8143898 DOI: 10.1002/jnr.24640] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 11/08/2022]
Abstract
Heroin, a mu agonist, acts through the mu opioid receptor. The mu opioid receptor gene, OPRM1, undergoes extensive alternative splicing, creating an array of splice variants that are conserved from rodent to humans. Increasing evidence suggests that these OPRM1 splice variants are pharmacologically important in mediating various actions of mu opioids, including analgesia, tolerance, physical dependence, rewarding behavior, as well as addiction. In the present study, we examine expression of the OPRM1 splice variant mRNAs in the medial prefrontal cortex (mPFC), one of the major brain regions involved in decision-making and drug-seeking behaviors, of male human heroin abusers and male rats that developed stable heroin-seeking behavior using an intravenous heroin self-administration (SA) model. The results show similar expression profiles among multiple OPRM1 splice variants in both human control subjects and saline control rats, illustrating conservation of OPRM1 alternative splicing from rodent to humans. Moreover, the expressions of several OPRM1 splice variant mRNAs were dysregulated in the postmortem mPFCs from heroin abusers compared to the control subjects. Similar patterns were observed in the rat heroin SA model. These findings suggest potential roles of the OPRM1 splice variants in heroin addiction that could be mechanistically explored using the rat heroin SA model.
Collapse
Affiliation(s)
- Taylor G Brown
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Jin Xu
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Yasmin L Hurd
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ying-Xian Pan
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
45
|
Graham DP, Harding MJ, Nielsen DA. Pharmacogenetics of Addiction Therapy. Methods Mol Biol 2022; 2547:437-490. [PMID: 36068473 DOI: 10.1007/978-1-0716-2573-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Drug addiction is a serious relapsing disease that has high costs to society and to the individual addicts. Treatment of these addictions is still in its nascency, with only a few examples of successful therapies. Therapeutic response depends upon genetic, biological, social, and environmental components. A role for genetic makeup in the response to treatment has been shown for several addiction pharmacotherapies with response to treatment based on individual genetic makeup. In this chapter, we will discuss the role of genetics in pharmacotherapies, specifically for cocaine, alcohol, and opioid dependences. The continued elucidation of the role of genetics should aid in the development of new treatments and increase the efficacy of existing treatments.
Collapse
Affiliation(s)
- David P Graham
- Michael E. DeBakey Veterans Affairs Medical Center, and the Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Mark J Harding
- Michael E. DeBakey Veterans Affairs Medical Center, and the Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - David A Nielsen
- Michael E. DeBakey Veterans Affairs Medical Center, and the Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
46
|
Kawakami K, Miyasaka T, Nakamura Y, Metoki H, Miyata S, Sato M, Sora I, Yamauchi K, Kawakami K, Blendy JA, Kawano T, Shimokawa H, Takayanagi M, Ohno I, Takahashi T. The A118G single-nucleotide polymorphism in OPRM1 is a risk factor for asthma severity. Allergol Int 2022; 71:55-65. [PMID: 34688555 DOI: 10.1016/j.alit.2021.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Although population studies have implicated emotional burden in asthma severity, the underlying genetic risk factors are not completely understood. We aimed to evaluate the genetic influence of a functional single-nucleotide polymorphism (SNP) in the stress-related μ-opioid receptor gene (OPRM1; A118G SNP, rs1799971) on asthma severity. METHODS We initially assessed disease severity in asthmatic outpatients carrying A118G. Using an ovalbumin-induced experimental asthma rodent model harboring the functionally equivalent SNP, we investigated the mechanism by which this SNP influences the allergic immune response. RESULTS Among 292 outpatients, 168 underwent airway hyperresponsiveness (AHR) to methacholine testing. Compared with patients carrying the AA and AG genotypes, those carrying the GG genotype exhibited enhanced AHR. The stress levels were presumed to be moderate among patients and were comparable among genotypes. Compared with Oprm1 AA mice, GG mice demonstrated aggravated asthma-related features and increased pulmonary interleukin-4+CD4+ effector and effector memory T cells under everyday life stress conditions. Intraperitoneal naloxone methiodide injection reduced effector CD4+ T cell elevation associated with increased eosinophil numbers in bronchoalveolar lavage fluid of GG mice to the levels in AA mice, suggesting that elevated Th2 cell generation in the bronchial lymph node (BLN) of GG mice induces enhanced eosinophilic inflammation. CONCLUSIONS Without forced stress exposure, patients with asthma carrying the OPRM1 GG genotype exhibit enhanced AHR, attributable to enhanced Th2 cell differentiation in the regional lymph node. Further research is necessary to elucidate the role of the OPRM1 A118G genotype in the Th2 cell differentiation pathway in the BLN.
Collapse
Affiliation(s)
- Kaori Kawakami
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tomomitsu Miyasaka
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
| | - Yutaka Nakamura
- Division of Pulmonary Medicine, Allergy, and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Hirohito Metoki
- Division of Public Health, Hygiene and Epidemiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Satoshi Miyata
- Teikyo University Graduate School of Public Health, Tokyo, Japan
| | - Miki Sato
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ichiro Sora
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kohei Yamauchi
- Division of Pulmonary Medicine, Allergy, and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Kazuyoshi Kawakami
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Julie A Blendy
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Medical School, Philadelphia, PA, USA
| | - Tasuku Kawano
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Motoaki Takayanagi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Isao Ohno
- Center for Medical Education, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tomoko Takahashi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
47
|
Blackwood CA, Cadet JL. Epigenetic and Genetic Factors Associated With Opioid Use Disorder: Are These Relevant to African American Populations. Front Pharmacol 2021; 12:798362. [PMID: 35002733 PMCID: PMC8727544 DOI: 10.3389/fphar.2021.798362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/07/2021] [Indexed: 01/02/2023] Open
Abstract
In the United States, the number of people suffering from opioid use disorder has skyrocketed in all populations. Nevertheless, observations of racial disparities amongst opioid overdose deaths have recently been described. Opioid use disorder is characterized by compulsive drug consumption followed by periods of withdrawal and recurrent relapses while patients are participating in treatment programs. Similar to other rewarding substances, exposure to opioid drugs is accompanied by epigenetic changes in the brain. In addition, genetic factors that are understudied in some racial groups may also impact the clinical manifestations of opioid use disorder. These studies are important because genetic factors and epigenetic alterations may also influence responses to pharmacological therapeutic approaches. Thus, this mini-review seeks to briefly summarize what is known about the genetic bases of opioid use disorder in African Americans.
Collapse
Affiliation(s)
- Christopher A. Blackwood
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, United States
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, United States
| |
Collapse
|
48
|
Veerappa A, Pendyala G, Guda C. A systems omics-based approach to decode substance use disorders and neuroadaptations. Neurosci Biobehav Rev 2021; 130:61-80. [PMID: 34411560 PMCID: PMC8511293 DOI: 10.1016/j.neubiorev.2021.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/23/2021] [Accepted: 08/14/2021] [Indexed: 11/15/2022]
Abstract
Substance use disorders (SUDs) are a group of neuropsychiatric conditions manifesting due to excessive dependence on potential drugs of abuse such as psychostimulants, opioids including prescription opioids, alcohol, inhalants, etc. Experimental studies have generated enormous data in the area of SUDs, but outcomes from such data have remained largely fragmented. In this review, we attempt to coalesce these data points providing an important first step towards our understanding of the etiology of SUDs. We propose and describe a 'core addictome' pathway that behaves central to all SUDs. Besides, we also have made some notable observations paving way for several hypotheses; MECP2 behaves as a master switch during substance use; five distinct gene clusters were identified based on respective substance addiction; a central cluster of genes serves as a hub of the addiction pathway connecting all other substance addiction clusters. In addition to describing these findings, we have emphasized the importance of some candidate genes that are of substantial interest for further investigation and serve as high-value targets for translational efforts.
Collapse
Affiliation(s)
- Avinash Veerappa
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Gurudutt Pendyala
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Center for Biomedical Informatics Research and Innovation, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
49
|
Swinford-Jackson SE, O'Brien CP, Kenny PJ, Vanderschuren LJMJ, Unterwald EM, Pierce RC. The Persistent Challenge of Developing Addiction Pharmacotherapies. Cold Spring Harb Perspect Med 2021; 11:a040311. [PMID: 32601131 PMCID: PMC8559539 DOI: 10.1101/cshperspect.a040311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
There are currently effective Food and Drug Administration (FDA)-approved therapies for alcohol, nicotine, and opioid use disorders. This article will review the development of eight compounds used in the treatment of drug addiction with an emphasis on pharmacological mechanisms and the utility of preclinical animal models of addiction in therapeutic development. In contrast to these successes, animal research has identified a number of promising medications for the treatment of psychostimulant use disorder, none of which have proven to be clinically effective. A specific example of an apparently promising pharmacotherapeutic for cocaine that failed clinically will be examined to determine whether this truly represents a challenge to the predictive validity of current models of cocaine addiction. In addition, the development of promising cocaine use disorder therapeutics derived from animal research will be reviewed, with some discussion regarding how preclinical studies might be modified to better inform clinical outcomes.
Collapse
Affiliation(s)
- Sarah E Swinford-Jackson
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Charles P O'Brien
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Paul J Kenny
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Louk J M J Vanderschuren
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, 3584 CM, Utrecht, The Netherlands
| | - Ellen M Unterwald
- Center for Substance Abuse Research and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, USA
| | - R Christopher Pierce
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
50
|
Montero AA, Vasconcelos SRPD. [Genetics and pain: approach and implications in clinical practice]. Semergen 2021; 47:431-433. [PMID: 34696866 DOI: 10.1016/j.semerg.2021.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 11/27/2022]
Affiliation(s)
- A Alcántara Montero
- Centro de Salud Manuel Encinas. Consultorio de Malpartida de Cáceres, Cáceres, España; Miembro del Grupo de Trabajo de Dolor y Cuidados Paliativos de SEMERGEN.
| | | |
Collapse
|