Skip to main content

MINI REVIEW article

Front. Pharmacol., 22 December 2021
Sec. Neuropharmacology
This article is part of the Research Topic Unraveling Vulnerability Factors in Addiction Drug Use and Potential Treatments View all 20 articles

Epigenetic and Genetic Factors Associated With Opioid Use Disorder: Are These Relevant to African American Populations

  • Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, United States

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.

Introduction

Opioid overdose is one of the leading causes of deaths in the United States (U.S.) (Jalal et al., 2018). The over-prescription of opioids and the illicit use of these drugs are amongst the factors that have contributed to the rise in opioid overdoses and the development of opioid use disorder (OUD) (CDC, 2020; Blackwood and Cadet, 2021b; Mattson et al., 2021). OUD is a chronic relapsing neuropsychiatric disorder characterized by compulsive drug intake despite negative life consequences (DSM-V, 2013). Importantly, there are reported racial differences in people suffering from OUD (Terry et al., 2008; Nielsen et al., 2010; Xu et al., 2018). In addition, certain African American populations are reported to be dying from opioid overdoses at higher rates than other ethnic groups (Carlesso and Kara, 2019; Chau, 2020; DeLaquil, 2020; Furr-Holden et al., 2021). It is therefore important to understand the potential causes for these observations of differential overdose rates for prevention purposes.

Opioid drugs have their biological effects by stimulating opioid receptors that are members of the G protein coupled receptor (GPCR) family (Shippenberg et al., 2008; Pasternak and Pan, 2013; Gendron et al., 2016). These receptors include mu(μ)-, delta(Δ)-, and kappa(Κ)-opioid receptors that can form homo- and heterodimeric complexes and transduce intercellular signals through various cellular pathways (Fujita et al., 2014; Bruchas and Roth, 2016). Among these, the μ-opioid receptor appears to be more relevant to addictive processes (Bossert et al., 2018; Blackwood et al., 2019) and is the main target for FDA-approved drugs used to treat OUD (Schuckit, 2016).

The μ-opioid receptor is encoded by the OPRM1 gene (Chen et al., 1993; Thompson et al., 1993). The receptor is expressed throughout the brain (Hirvonen et al., 2009; Johansson et al., 2019) and has high affinity to endogenous opiates including ß-endorphin (Zadina et al., 1997). Stimulation of μ-opioid receptor is responsible, in part, for opioid-induced euphoria and rewarding effects (Wang, 2019). Repeated stimulation of the μ-opioid receptor is also mainly responsible for the development of tolerance and physical dependence on opioid drugs (Kieffer and Evans, 2002). In clinical settings, various pain syndromes are treated with μ-opioid receptor agonists (Basbaum and Fields, 1984; Pasternak, 1993). Therefore, factors that interfere with the pharmacokinetics and signaling of the μ-opioid receptor could impact the treatment of pains syndrome and/or the manifestation of OUD in various ethnic populations.

Here, we provide a short review of potential genetic and epigenetic factors that impact the μ-opioid receptor gene. These include mutations reported in the μ-opioid receptor gene and some epigenetic factors including DNA methylation and transgenerational epigenetic inheritance that might impact gene expression in the brain. Finally, we consider the possibility that these might impact clinical course of OUD in African American populations.

Single Nucleotide Polymorphisms

Genetic variations due to single nucleotide polymorphisms (SNPs) are factors that can influence susceptibility to substance use disorders (SUDs) (Oslin et al., 2003; Haerian and Haerian, 2013). Most SNPs are not necessarily associated with any discernible functional changes in mRNA expression or protein functions. However, some SNPs can result in amino-acid substitutions that change the functionality of proteins that can result in susceptibility to brain diseases or adverse consequences during exposure to stressful environmental stimuli (Shi et al., 2011; Lee et al., 2017).

SNPs Identified in the μ-Opioid Receptor Gene

Several groups of investigators have reported on potential links between OUD and SNPs identified in the human μ-opioid receptor gene (Bond et al., 1998; Zhang et al., 2005). These SNPs include C17T, G24A, G799A, G942A, and A118G (Bond et al., 1998), with the A118G variant being the most commonly reported. This SNP is located at position 118 (A118G) and corresponds to an amino-acid conversion from asparagine to aspartate at position 40 of the N terminus site of the receptor protein (Bond et al., 1998). Interestingly, Bond et al. (1998), reported that the A118G variant showed greater affinity for ß-endorphin. In addition, activation of the A118G variant is much more potent at the G-protein-coupled protein potassium ion channels than the normal variant (Bond et al., 1998). Of further clinical relevance, the A118G variant is an important mediator of endocytosis and desensitization of the human μ-opioid receptor (Beyer et al., 2004), implicating it in the development of greater tolerance in individual patients who use or misuse opioid drugs. Postmortem tissues that have revealed decreased mRNA expression and protein levels of the human μ-opioid receptor in humans with the A118G variant (Zhang et al., 2005) further implicate them in the clinical course of OUD patients.

Potential Implications of A118G in African American Patients With OUD

It has been reported that the occurrence of the A118G variant may vary across ethnic groups (Bond et al., 1998; Hastie et al., 2012; Abijo et al., 2020). Specifically, allele frequencies of the A118G variant have been investigated in healthy and opioid-exposed African Americans (Bond et al., 1998; Hastie et al., 2012). Bond et al. (1998) used heroin-exposed and non-drug exposed subjects found an overall allelic frequency (A/G and G/G) of 3.3% in African Americans compared to 21.2 and 25.4% in Whites and Hispanics, respectively. A subsequent study by Hastie et al. (2012) who included healthy young adults in their paper documented that the frequency of the A118G variant was less common in African Americans (7.4%) compared to Whites (28.7%) and Hispanics (27.8%). The presence of A118G polymorphisms in African Americans and other ethnic groups might affect the binding affinity of the μ-opioid receptor. The changes in binding affinity might hamper pain perception, reduce response to analgesic drugs, and increase self-administration of opioid drugs (Beyer et al., 2004; Janicki et al., 2006; Sia et al., 2008).

A118G polymorphism removes a highly conserved N-glycosylation site in protein’s extracellular domain (Beyer et al., 2004) that may hamper pain perception in chronic diseases (Janicki et al., 2006), reduce response towards analgesic drugs (Oertel et al., 2006), and tend to increase administration of opioids (Sia et al., 2008).

DNA Methylation in Eukaryotic Cells

Repeated exposure to drugs that lead to SUD in some individuals is related, in part, to neuroadaptive epigenetic alterations that occur in the brains of exposed individuals (Robison and Nestler, 2011; Cadet, 2016; Cadet and Jayanthi, 2021). Epigenetic events occur through DNA methylation, chromatin remodeling, non-coding RNA, and histone modifications (Robison and Nestler, 2011). The next paragraphs focus on the role of DNA methylation in OUD because DNA methylation plays an important role in the transcription of the μ-opioid receptor gene (Figure 1) (Hwang et al., 2007; Chidambaran et al., 2017).

FIGURE 1
www.frontiersin.org

FIGURE 1. Opioid-induced DNA methylation of mu opioid receptor gene. Agents such as opioids have been shown to cause epigenetic alterations in the brain. This cartoon illustrates how exposure to a mu opioid agonist can cause hypermethylation at CPG sites located in the promoter regions of the mu opioid receptor gene. DNA methylation is followed by the recruitment of co-repressors including HDAC1, DNMTs, and/or MBD proteins (MeCP2). Together these protein form complexes that can repress gene transcription repression by preventing binding of transcription factors on the DNA promoters or relevant transcription sites of relevant genes.

DNA methylation refers to the addition of methyl groups to cytosine residue. This reaction is catalyzed by the enzymes, DNA methyltransferases (DNMTs) (Kinney and Pradhan, 2011; Smith and Meissner, 2013). Changes in DNA methylation can occur in enhancers and promoters of genes (Kinney and Pradhan, 2011; Smith and Meissner, 2013). DNA methylation is required for normal development in humans (Kim et al., 2009; Smith and Meissner, 2013). Changes in DNA methylation may lead to repression of gene expression through the recruitment of histone deacetylases (HDACs), histone methyl/lysine-transferases (HMATs/KMTs), as well as DNA methyl-binding domain proteins (MBDs) methyl-CpG binding protein 2 (MeCP2) (Hyun et al., 2017; Voss and Thomas, 2018). Alterations in DNA methylation are influenced by environmental stress and chemical exposure. DNA methylation plays important roles in the pathobiology of cancer and neurodevelopmental disorders (Robertson and Wolffe, 2000).

DNA Methylation and μ-Opioid Receptors

Alterations in DNA methylation have been reported after chronic exposure to several rewarding drugs including opioids (Hwang et al., 2007; Host et al., 2011; Manzardo et al., 2012; Pol Bodetto et al., 2013; Chidambaran et al., 2017; Jayanthi et al., 2020). Specifically, Hwang et al. (2007) had reported that increased DNA methylation of CpG sites in the promoter regions of the μ-opioid receptor gene led to increased binding of MeCP2 followed by recruitment of the repressors, histone deacetylase 1 (HDAC1) and SIN3 transcription regulator family member A (mSin3A), thus leading to downregulation of the μ-opioid receptor gene. Their findings and interpretation are consistent with observations that MeCP2 can negatively regulate the expression of the μ-opioid receptor gene (Lu et al., 2009; Garcia-Concejo et al., 2016).

DNA Methylation and Opioid Use Disorder

Aberrant alterations in DNA methylation have been reported to be associated with various neuropsychiatric diseases including heroin use disorder (Nielsen et al., 2008; Cadet, 2016; Wang et al., 2016; Cadet and Jayanthi, 2021). For example, Nielsen et al. (2008) reported that DNA taken from lymphocytes of patients suffering from heroin use disorder showed increased levels of DNA methylation. Similarly, Xu et al. (2018) reported DNA hypermethylation in former patients suffering from heroin use disorder compared to healthy controls who had no history of opioid consumption. The results of these two papers are consistent with those of other investigators who have found that patients suffering from heroin use disorder showed altered DNA methylation at CpG sites located in the promoter region of the μ-opioid receptor (Chorbov et al., 2011; Doehring et al., 2013; Ebrahimi et al., 2018). Specifically, Chorbov et al. (2011) reported that former patients suffering from heroin use disorder stabilized in methadone treatment showed increased DNA methylation in two CPG sites at +182 and +186 loci of the promoted of the μ-opioid receptor. In addition, Doehring et al. (2013) documented increased DNA methylation in one CpG site at +136 loci of the receptor promoter in heroin-dependent patients and in opioid-treated pain patients in comparison to the non-opioid exposed individuals (Doehring et al., 2013). Taken together, these results implicate a role of DNA methylation of the opioid receptor gene in OUD.

DNA Methylation and African Americans

Although limited, it has been suggested that there are differential changes in DNA methylation levels across individuals from various racial groups exposed to opioids (Nielsen et al., 2009; Nielsen et al., 2010). Nielsen et al. (2009) had initially reported that hypermethylation at CpG sites located in the promoter of the μ-opioid receptor was linked to long-term heroin consumption. Subsequently, they observed that African American individual patients showed higher levels of DNA methylation upstream of the promoter region of the μ-opioid receptor in comparison to Hispanic or White patients (Nielsen et al., 2010). These observations need to be replicated in much larger patient populations before any rigorous interpretations can be made with confidence.

Intergenerational and Transgenerational Epigenetic Inheritance

Opioid exposure is thought to be associated with changes in a germline, which may be transmitted to subsequent generations in a process called intergenerational or transgenerational epigenetic inheritance (Cicero et al., 1995; Sarkaki et al., 2008; Byrnes et al., 2012; Byrnes et al., 2013; Vassoler et al., 2017; Toorie et al., 2021). Intergenerational and transgenerational epigenetic inheritance refers to phenotypic variation that does not stem from variations in DNA base sequences that are transmitted through the germline to the immediate offspring even in the absence of direct opioid exposure (Vassoler et al., 2014; Odegaard et al., 2020). Some of these ideas are illustrated in Figure 2.

FIGURE 2
www.frontiersin.org

FIGURE 2. Transgenerational and Multigenerational epigenetic inheritance of opioid exposure. The ability to transmit epigenetic changes from one generation to the next in experiments done in animal models. There is very little direct evidence of similar occurrences in humans. Opioid use by mothers (or fathers) might lead to permanent changes in the DNA found in their ovaries (or sperm) that are transmissible to later generations even in the absent of direct drug exposure (transgenerational epigenetic inheritance). Multigenerational epigenetic inheritance coincident direct exposure of multiple generations to an environmental factors promoting alterations in the multiple generations exposed.

In an interesting study, adolescent female rats injected with increasing dosages of morphine were mated with drug-free males (F0 animals) (Byrnes et al., 2013). The F1 and F2 offsprings that derived from the F0 female were then investigated for locomotor defects after injection with a dopamine D2 receptor (D2R) activator, quinpirole. Repeated administration of quinpirole in the F1 and F2 progenies reduced locomotor sensitization in activity testing chambers (Byrnes et al., 2013). In the same study, the F1 and F2 progenies showed increased expression of D2R and Κ-opioid receptors in the nucleus accumbens. Using a similar model with morphine, F1 offspring from the morphine-exposed female showed decreased anxiety-like behavior in open field activity experiments and increased sensitivity to opioid rewarding effects (Byrnes et al., 2011).

In another interesting study, performed by Vassoler et al. (2017), females adolescent rats exposed to morphine were bred with drug-naïve males. In this study, F1 and F2 offsprings from the maternal line were found to have lower levels of morphine self-administration and reduced relapse-like behavior. Additionally, they showed altered expression of genes associated with synaptic plasticity in the nucleus accumbens (Vassoler et al., 2017). A study performed by Odegaard et al. (2020) found that pregnant mothers exposed to oxycodone showed developmental impairments that were displayed in multiple generations. These findings illustrated the potential transgenerational and multigenerational influences of opioids exposure in females.

General Summary and Conclusion

The prevalence of OUD and its associated consequences including overdose deaths have increased in recent years. Unfortunately, African Americans have been reported to have suffered some of the largest increases in opioid-related overdose deaths (Patel et al., 2021). The potential ramifications of these changes in the course of the COVID-19 infection has also been discussed (Blackwood and Cadet, 2021a). Some of these complications may be related to the lack of access of African American individual patients to psychiatric care that has been shown to be an area of major racial inequities (Hall et al., 2021; Stahler et al., 2021). This discussion suggests the need to increase available resources to increase access to treatment programs by African Americans who seem to be suffering from the brunt of the disasters associated with the opioid epidemic in the United States.

It needs to be further commented that more expansive genetic and epigenetic studies are needed in order to compare individual and group racial differences in the susceptibility to or resistance against OUD. This statement is based on some initial studies that have reported differences in genetic and epigenetic markers across various ethnic groups (Nielsen et al., 2010; Abijo et al., 2020). For example, it will be important to investigate the potential connections between the A118G variant and the clinical course of OUD in populations that include large numbers of patients and non-patients from various ethnic American groups. So far, it is very obvious that most of these genetic studies have focused on white populations and have neglected the African American communities in the United States. Without engaging these populations, the racial inequities in diagnosis, treatment, and mortality will continue to rise. Similarly, the initial findings of differences in DNA methylation of the opioid receptor gene need to be investigated further in similar large population of controls and patients from various ethnic groups. Without these studies, it is possible that prevention approaches that treat all populations based on findings in white populations will fail. The suggested approaches are also relevant to the practice of precision medicine that will take the treatment of SUD into the next decades.

Author Contributions

CB drafted, created illustrations, and wrote the manuscript. JC wrote, conceptualized, reviewed and edited the contents of the paper. Both authors approved the final draft of the manuscript.

Funding

This work is supported by funds of the Intramural Research Program of the DHHS/NIH/NIDA.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abijo, T., Blum, K., and Gondré-Lewis, M. C. (2020). Neuropharmacological and Neurogenetic Correlates of Opioid Use Disorder (OUD) As a Function of Ethnicity: Relevance to Precision Addiction Medicine. Curr Neuropharmacol 18 (7), 578–595. doi:10.2174/1570159X17666191118125702

PubMed Abstract | CrossRef Full Text | Google Scholar

Basbaum, A. I., and Fields, H. L. (1984). Endogenous Pain Control Systems: Brainstem Spinal Pathways and Endorphin Circuitry. Annu Rev Neurosci 7, 309–338. doi:10.1146/annurev.ne.07.030184.001521

PubMed Abstract | CrossRef Full Text | Google Scholar

Beyer, A., Koch, T., Schröder, H., Schulz, S., and Höllt, V. (2004). Effect of the A118G Polymorphism on Binding Affinity, Potency and Agonist-Mediated Endocytosis, Desensitization, and Resensitization of the Human Mu-Opioid Receptor. J Neurochem 89 (3), 553–560. doi:10.1111/j.1471-4159.2004.02340.x

CrossRef Full Text | Google Scholar

Blackwood, C. A., and Cadet, J. L. (2021a). COVID-19 Pandemic and Fentanyl Use Disorder in African Americans. Front Neurosci 15, 707386. doi:10.3389/fnins.2021.707386

PubMed Abstract | CrossRef Full Text | Google Scholar

Blackwood, C. A., Hoerle, R., Leary, M., Schroeder, J., Job, M. O., McCoy, M. T., et al. (2019). Molecular Adaptations in the Rat Dorsal Striatum and Hippocampus Following Abstinence-Induced Incubation of Drug Seeking After Escalated Oxycodone Self-Administration. Mol Neurobiol 56 (5), 3603–3615. doi:10.1007/s12035-018-1318-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Blackwood, C. A., and Cadet, J. L. (2021b). The Molecular Neurobiology and Neuropathology of Opioid Use Disorder. Current Research in Neurobiology 2, 100023. doi:10.1016/j.crneur.2021.100023

CrossRef Full Text | Google Scholar

Bond, C., LaForge, K. S., Tian, M., Melia, D., Zhang, S., Borg, L., et al. (1998). Single-nucleotide Polymorphism in the Human Mu Opioid Receptor Gene Alters Beta-Endorphin Binding and Activity: Possible Implications for Opiate Addiction. Proc Natl Acad Sci U S A 95 (16), 9608–9613. doi:10.1073/pnas.95.16.9608

PubMed Abstract | CrossRef Full Text | Google Scholar

Bossert, J. M., Hoots, J. K., Fredriksson, I., Adhikary, S., Zhang, M., Venniro, M., et al. (2018). Role of Mu, but Not delta or Kappa, Opioid Receptors in Context‐induced Reinstatement of Oxycodone Seeking. Eur J Neurosci 50, 2075–2085. doi:10.1111/ejn.13955

CrossRef Full Text | Google Scholar

Bruchas, M. R., and Roth, B. L. (2016). New Technologies for Elucidating Opioid Receptor Function. Trends Pharmacol Sci 37 (4), 279–289. doi:10.1016/j.tips.2016.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Byrnes, J. J., Babb, J. A., Scanlan, V. F., and Byrnes, E. M. (2011). Adolescent Opioid Exposure in Female Rats: Transgenerational Effects on Morphine Analgesia and Anxiety-like Behavior in Adult Offspring. Behav Brain Res 218 (1), 200–205. doi:10.1016/j.bbr.2010.11.059

PubMed Abstract | CrossRef Full Text | Google Scholar

Byrnes, J. J., Johnson, N. L., Carini, L. M., and Byrnes, E. M. (2013). Multigenerational Effects of Adolescent Morphine Exposure on Dopamine D2 Receptor Function. Psychopharmacology (Berl) 227 (2), 263–272. doi:10.1007/s00213-012-2960-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Byrnes, J. J., Johnson, N. L., Schenk, M. E., and Byrnes, E. M. (2012). Cannabinoid Exposure in Adolescent Female Rats Induces Transgenerational Effects on Morphine Conditioned Place Preference in Male Offspring. J Psychopharmacol 26 (10), 1348–1354. doi:10.1177/0269881112443745

CrossRef Full Text | Google Scholar

Cadet, J. L. (2016). Epigenetics of Stress, Addiction, and Resilience: Therapeutic Implications. Mol Neurobiol 53 (1), 545–560. doi:10.1007/s12035-014-9040-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Cadet, J. L., and Jayanthi, S. (2021). Epigenetics of Addiction. Neurochem Int 147, 105069. doi:10.1016/j.neuint.2021.105069

PubMed Abstract | CrossRef Full Text | Google Scholar

Carlesso, K. J., and Kara, J. (2019). Best of 2019: Blacks Dying Form Fentanyl at Same Rate as Whites for the First Time. The CT Mirror. Available at: https://ctmirror.org/2019/12/27/best-of-2019-blacks-dying-from-fentanyl-at-same-rate-as-whites-for-first-time/.

Google Scholar

CDC (2020). Increase in Fatal Drug Overdoses across the United States Driven by Synthetic Opioids before and during the COVID-19 Pandemic. Health Alert Network. Available at: https://emergency.cdc.gov/han/2020/han00438.asp.

Google Scholar

Chau, V. (2020). Substance Abuse and Mental Health Services Administration: The Opioid Crisis and the Black/African American Population: An Urgent Issue. Available at: https://store.samhsa.gov/product/The-Opioid-Crisis-and-the-Black-African-American-Population-An-Urgent-Issue/PEP20-05-02-001.

Google Scholar

Chen, Y., Mestek, A., Liu, J., Hurley, J. A., and Yu, L. (1993). Molecular Cloning and Functional Expression of a Mu-Opioid Receptor from Rat Brain. Mol Pharmacol 44 (1), 8–12. Available at: https://www.ncbi.nlm.nih.gov/pubmed/8393525.

PubMed Abstract | Google Scholar

Chidambaran, V., Zhang, X., Martin, L. J., Ding, L., Weirauch, M. T., Geisler, K., et al. (2017). DNA Methylation at the Mu-1 Opioid Receptor Gene (OPRM1) Promoter Predicts Preoperative, Acute, and Chronic Postsurgical Pain after Spine Fusion. Pharmgenomics Pers Med 10, 157–168. doi:10.2147/PGPM.S132691

PubMed Abstract | CrossRef Full Text | Google Scholar

Chorbov, V. M., Todorov, A. A., Lynskey, M. T., and Cicero, T. J. (2011). Elevated Levels of DNA Methylation at the OPRM1 Promoter in Blood and Sperm from Male Opioid Addicts. J Opioid Manag 7 (4), 258–264. doi:10.5055/jom.2011.0067

CrossRef Full Text | Google Scholar

Cicero, T. J., Nock, B., O'Connor, L., Adams, M., and Meyer, E. R. (1995). Adverse Effects of Paternal Opiate Exposure on Offspring Development and Sensitivity to Morphine-Induced Analgesia. J Pharmacol Exp Ther 273 (1), 386–392. Available at: https://www.ncbi.nlm.nih.gov/pubmed/7714793.

Google Scholar

DeLaquil, M. (2020). Differences in Rates of Drug Overdose Deaths by Race. Available at: https://www.health.state.mn.us/communities/opioids/documents/raceratedisparity2019prelimfinal.pdf.

Google Scholar

Doehring, A., Oertel, B. G., Sittl, R., and Lötsch, J. (2013). Chronic Opioid Use Is Associated with Increased DNA Methylation Correlating with Increased Clinical Pain. Pain 154 (1), 15–23. doi:10.1016/j.pain.2012.06.011

PubMed Abstract | CrossRef Full Text | Google Scholar

DSM-V.(2013). Diagnostic and Statistical Manual of Mental Disorders. Washington, DC: American Psychiatric Association.

Google Scholar

Ebrahimi, G., Asadikaram, G., Akbari, H., Nematollahi, M. H., Abolhassani, M., Shahabinejad, G., et al. (2018). Elevated Levels of DNA Methylation at the OPRM1 Promoter Region in Men with Opioid Use Disorder. Am J Drug Alcohol. Abuse 44 (2), 193–199. doi:10.1080/00952990.2016.1275659

CrossRef Full Text | Google Scholar

Fujita, W., Gomes, I., and Devi, L. A. (2014). Revolution in GPCR Signalling: Opioid Receptor Heteromers as Novel Therapeutic Targets: IUPHAR Review 10. Br J Pharmacol 171 (18), 4155–4176. doi:10.1111/bph.12798

CrossRef Full Text | Google Scholar

Furr-Holden, D., Milam, A. J., Wang, L., and Sadler, R. (2021). African Americans Now Outpace Whites in Opioid-Involved Overdose Deaths: a Comparison of Temporal Trends from 1999 to 2018. Addiction 116 (3), 677–683. doi:10.1111/add.15233

PubMed Abstract | CrossRef Full Text | Google Scholar

Garcia-Concejo, A., Jimenez-Gonzalez, A., and Rodríguez, R. E. (2016). μ Opioid Receptor Expression after Morphine Administration Is Regulated by miR-212/132 Cluster. PLoS One 11 (7), e0157806. doi:10.1371/journal.pone.0157806

PubMed Abstract | CrossRef Full Text | Google Scholar

Gendron, L., Cahill, C. M., von Zastrow, M., Schiller, P. W., and Pineyro, G. (2016). Molecular Pharmacology of δ-Opioid Receptors. Pharmacol Rev 68 (3), 631–700. doi:10.1124/pr.114.008979

PubMed Abstract | CrossRef Full Text | Google Scholar

Haerian, B. S., and Haerian, M. S. (2013). OPRM1 Rs1799971 Polymorphism and Opioid Dependence: Evidence from a Meta-Analysis. Pharmacogenomics 14 (7), 813–824. doi:10.2217/pgs.13.57

PubMed Abstract | CrossRef Full Text | Google Scholar

Hall, G. C. N., Berkman, E. T., Zane, N. W., Leong, F. T. L., Hwang, W. C., Nezu, A. M., et al. (2021). Reducing Mental Health Disparities by Increasing the Personal Relevance of Interventions. Am Psychol 76 (1), 91–103. doi:10.1037/amp0000616

PubMed Abstract | CrossRef Full Text | Google Scholar

Hastie, B. A., Riley, J. L., Kaplan, L., Herrera, D. G., Campbell, C. M., Virtusio, K., et al. (2012). Ethnicity Interacts with the OPRM1 Gene in Experimental Pain Sensitivity. Pain 153 (8), 1610–1619. doi:10.1016/j.pain.2012.03.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirvonen, J., Aalto, S., Hagelberg, N., Maksimow, A., Ingman, K., Oikonen, V., et al. (2009). Measurement of central Mu-Opioid Receptor Binding In Vivo with PET and [11C]carfentanil: a Test-Retest Study in Healthy Subjects. Eur J Nucl Med Mol Imaging 36 (2), 275–286. doi:10.1007/s00259-008-0935-6

CrossRef Full Text | Google Scholar

Host, L., Dietrich, J. B., Carouge, D., Aunis, D., and Zwiller, J. (2011). Cocaine Self-Administration Alters the Expression of Chromatin-Remodelling Proteins; Modulation by Histone Deacetylase Inhibition. J Psychopharmacol 25 (2), 222–229. doi:10.1177/0269881109348173

CrossRef Full Text | Google Scholar

Hwang, C. K., Song, K. Y., Kim, C. S., Choi, H. S., Guo, X. H., Law, P. Y., et al. (2007). Evidence of Endogenous Mu Opioid Receptor Regulation by Epigenetic Control of the Promoters. Mol Cell Biol 27 (13), 4720–4736. doi:10.1128/MCB.00073-07

PubMed Abstract | CrossRef Full Text | Google Scholar

Hyun, K., Jeon, J., Park, K., and Kim, J. (2017). Writing, Erasing and reading Histone Lysine Methylations. Exp Mol Med 49 (4), e324. doi:10.1038/emm.2017.11

PubMed Abstract | CrossRef Full Text | Google Scholar

Jalal, H., Buchanich, J. M., Roberts, M. S., Balmert, L. C., Zhang, K., and Burke, D. S. (2018). Changing Dynamics of the Drug Overdose Epidemic in the United States from 1979 through 2016. Science 361 (6408), 361. doi:10.1126/science.aau1184

CrossRef Full Text | Google Scholar

Janicki, P. K., Schuler, G., Francis, D., Bohr, A., Gordin, V., Jarzembowski, T., et al. (2006). A Genetic Association Study of the Functional A118G Polymorphism of the Human Mu-Opioid Receptor Gene in Patients with Acute and Chronic Pain. Anesth Analg 103 (4), 1011–1017. doi:10.1213/01.ane.0000231634.20341.88

PubMed Abstract | CrossRef Full Text | Google Scholar

Jayanthi, S., Torres, O. V., Ladenheim, B., and Cadet, J. L. (2020). A Single Prior Injection of Methamphetamine Enhances Methamphetamine Self-Administration (SA) and Blocks SA-Induced Changes in DNA Methylation and mRNA Expression of Potassium Channels in the Rat Nucleus Accumbens. Mol Neurobiol 57 (3), 1459–1472. doi:10.1007/s12035-019-01830-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Johansson, J., Hirvonen, J., Lovró, Z., Ekblad, L., Kaasinen, V., Rajasilta, O., et al. (2019). Intranasal Naloxone Rapidly Occupies Brain Mu-Opioid Receptors in Human Subjects. Neuropsychopharmacology 44 (9), 1667–1673. doi:10.1038/s41386-019-0368-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Kieffer, B. L., and Evans, C. J. (2002). Opioid Tolerance-In Search of the Holy Grail. Cell 108 (5), 587–590. doi:10.1016/s0092-8674(02)00666-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, J. K., Samaranayake, M., and Pradhan, S. (2009). Epigenetic Mechanisms in Mammals. Cell Mol Life Sci 66 (4), 596–612. doi:10.1007/s00018-008-8432-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Kinney, S. R., and Pradhan, S. (2011). Regulation of Expression and Activity of DNA (Cytosine-5) Methyltransferases in Mammalian Cells. Prog Mol Biol Transl Sci 101, 311–333. doi:10.1016/B978-0-12-387685-0.00009-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, K. H., Kim, T. W., Kang, J. H., Kim, J. S., Ahn, J. S., Kim, S. Y., et al. (2017). Efficacy and Safety of Controlled-Release Oxycodone/naloxone versus Controlled-Release Oxycodone in Korean Patients with Cancer-Related Pain: a Randomized Controlled Trial. Chin. J Cancer 36 (1), 74. doi:10.1186/s40880-017-0241-4

CrossRef Full Text | Google Scholar

Lu, M. C., Lai, T. Y., Hwang, J. M., Chen, H. T., Chang, S. H., Tsai, F. J., et al. (2009). Proliferation- and Migration-Enhancing Effects of Ginseng and Ginsenoside Rg1 through IGF-I- and FGF-2-Signaling Pathways on RSC96 Schwann Cells. Cell Biochem Funct 27 (4), 186–192. doi:10.1002/cbf.1554

PubMed Abstract | CrossRef Full Text | Google Scholar

Manzardo, A. M., Henkhaus, R. S., and Butler, M. G. (2012). Global DNA Promoter Methylation in Frontal Cortex of Alcoholics and Controls. Gene 498 (1), 5–12. doi:10.1016/j.gene.2012.01.096

PubMed Abstract | CrossRef Full Text | Google Scholar

Mattson, C. L., Tanz, L. J., Quinn, K., Kariisa, M., Patel, P., and Davis, N. L. (2021). Trends and Geographic Patterns in Drug and Synthetic Opioid Overdose Deaths - United States, 2013-2019. MMWR Morb Mortal Wkly Rep 70 (6), 202–207. doi:10.15585/mmwr.mm7006a4

PubMed Abstract | CrossRef Full Text | Google Scholar

Nielsen, D. A., Hamon, S., Yuferov, V., Jackson, C., Ho, A., Ott, J., et al. (2010). Ethnic Diversity of DNA Methylation in the OPRM1 Promoter Region in Lymphocytes of Heroin Addicts. Hum Genet 127 (6), 639–649. doi:10.1007/s00439-010-0807-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Nielsen, D. A., Ji, F., Yuferov, V., Ho, A., Chen, A., Levran, O., et al. (2008). Genotype Patterns that Contribute to Increased Risk for or protection from Developing Heroin Addiction. Mol Psychiatry 13 (4), 417–428. doi:10.1038/sj.mp.4002147

PubMed Abstract | CrossRef Full Text | Google Scholar

Odegaard, K. E., Schaal, V. L., Clark, A. R., Koul, S., Gowen, A., Sankarasubramani, J., et al. (2020). Characterization of the Intergenerational Impact of In Utero and Postnatal Oxycodone Exposure. Transl Psychiatry 10 (1), 329. doi:10.1038/s41398-020-01012-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Oertel, B. G., Schmidt, R., Schneider, A., Geisslinger, G., and Lötsch, J. (2006). The Mu-Opioid Receptor Gene Polymorphism 118A>G Depletes Alfentanil-Induced Analgesia and Protects against Respiratory Depression in Homozygous Carriers. Pharmacogenet Genomics 16, 625–636. doi:10.1097/01.fpc.0000220566.90466.a2

PubMed Abstract | CrossRef Full Text | Google Scholar

Oslin, D. W., Berrettini, W., Kranzler, H. R., Pettinati, H., Gelernter, J., Volpicelli, J. R., et al. (2003). A Functional Polymorphism of the Mu-Opioid Receptor Gene Is Associated with Naltrexone Response in Alcohol-dependent Patients. Neuropsychopharmacology 28 (8), 1546–1552. doi:10.1038/sj.npp.1300219

PubMed Abstract | CrossRef Full Text | Google Scholar

Pasternak, G. W., and Pan, Y. X. (2013). Mu Opioids and Their Receptors: Evolution of a Concept. Pharmacol Rev 65 (4), 1257–1317. doi:10.1124/pr.112.007138

PubMed Abstract | CrossRef Full Text | Google Scholar

Pasternak, G. W. (1993). Pharmacological Mechanisms of Opioid Analgesics. Clin Neuropharmacol 16 (1), 1–18. doi:10.1097/00002826-199302000-00001

PubMed Abstract | CrossRef Full Text | Google Scholar

Patel, I., Walter, L. A., and Li, L. (2021). Opioid Overdose Crises during the COVID-19 Pandemic: Implication of Health Disparities. Harm Reduct J 18 (1), 89. doi:10.1186/s12954-021-00534-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Pol Bodetto, S., Carouge, D., Fonteneau, M., Dietrich, J. B., Zwiller, J., and Anglard, P. (2013). Cocaine Represses Protein phosphatase-1Cβ through DNA Methylation and Methyl-CpG Binding Protein-2 Recruitment in Adult Rat Brain. Neuropharmacology 73, 31–40. doi:10.1016/j.neuropharm.2013.05.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Robertson, K. D., and Wolffe, A. P. (2000). DNA Methylation in Health and Disease. Nat Rev Genet 1 (1), 11–19. doi:10.1038/35049533

PubMed Abstract | CrossRef Full Text | Google Scholar

Robison, A. J., and Nestler, E. J. (2011). Transcriptional and Epigenetic Mechanisms of Addiction. Nat Rev Neurosci 12 (11), 623–637. doi:10.1038/nrn3111

PubMed Abstract | CrossRef Full Text | Google Scholar

Sarkaki, A., Assaei, R., Motamedi, F., Badavi, M., and Pajouhi, N. (2008). Effect of Parental Morphine Addiction on Hippocampal Long-Term Potentiation in Rats Offspring. Behav Brain Res 186 (1), 72–77. doi:10.1016/j.bbr.2007.07.041

PubMed Abstract | CrossRef Full Text | Google Scholar

Schuckit, M. A. (2016). Treatment of Opioid-Use Disorders. N Engl J Med 375 (16), 1596–1597. doi:10.1056/NEJMc1610830

CrossRef Full Text | Google Scholar

Shi, Y., Li, Z., Xu, Q., Wang, T., Li, T., Shen, J., et al. (2011). Common Variants on 8p12 and 1q24.2 Confer Risk of Schizophrenia. Nat Genet 43 (12), 1224–1227. doi:10.1038/ng.980

PubMed Abstract | CrossRef Full Text | Google Scholar

Shippenberg, T. S., LeFevour, A., and Chefer, V. I. (2008). Targeting Endogenous Mu- and delta-opioid Receptor Systems for the Treatment of Drug Addiction. CNS Neurol Disord Drug Targets 7 (5), 442–453. doi:10.2174/187152708786927813

PubMed Abstract | CrossRef Full Text | Google Scholar

Sia, A. T., Lim, Y., Lim, E. C., Goh, R. W., Law, H. Y., Landau, R., et al. (2008). A118G Single Nucleotide Polymorphism of Human Mu-Opioid Receptor Gene Influences Pain Perception and Patient-Controlled Intravenous Morphine Consumption after Intrathecal Morphine for Postcesarean Analgesia. Anesthesiology 109 (3), 520–526. doi:10.1097/ALN.0b013e318182af21

PubMed Abstract | CrossRef Full Text | Google Scholar

Smith, Z. D., and Meissner, A. (2013). DNA Methylation: Roles in Mammalian Development. Nat Rev Genet 14 (3), 204–220. doi:10.1038/nrg3354

PubMed Abstract | CrossRef Full Text | Google Scholar

Stahler, G. J., Mennis, J., and Baron, D. A. (2021). Racial/ethnic Disparities in the Use of Medications for Opioid Use Disorder (MOUD) and Their Effects on Residential Drug Treatment Outcomes in the US. Drug Alcohol Depend 226, 108849. doi:10.1016/j.drugalcdep.2021.108849

PubMed Abstract | CrossRef Full Text | Google Scholar

Terry, M. B., Ferris, J. S., Pilsner, R., Flom, J. D., Tehranifar, P., Santella, R. M., et al. (2008). Genomic DNA Methylation Among Women in a Multiethnic New York City Birth Cohort. Cancer Epidemiol Biomarkers Prev 17 (9), 2306–2310. doi:10.1158/1055-9965.EPI-08-0312

PubMed Abstract | CrossRef Full Text | Google Scholar

Thompson, R. C., Mansour, A., Akil, H., and Watson, S. J. (1993). Cloning and Pharmacological Characterization of a Rat Mu Opioid Receptor. Neuron 11 (5), 903–913. doi:10.1016/0896-6273(93)90120-g

PubMed Abstract | CrossRef Full Text | Google Scholar

Toorie, A. M., Vassoler, F. M., Qu, F., Schonhoff, C. M., Bradburn, S., Murgatroyd, C. A., et al. (2021). A History of Opioid Exposure in Females Increases the Risk of Metabolic Disorders in Their Future Male Offspring. Addict. Biol 26 (1), e12856. doi:10.1111/adb.12856

PubMed Abstract | CrossRef Full Text | Google Scholar

Vassoler, F. M., Byrnes, E. M., and Pierce, R. C. (2014). The Impact of Exposure to Addictive Drugs on Future Generations: Physiological and Behavioral Effects. Neuropharmacology 76 Pt B, 269–275. doi:10.1016/j.neuropharm.2013.06.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Vassoler, F. M., Oliver, D. J., Wyse, C., Blau, A., Shtutman, M., Turner, J. R., et al. (2017). Transgenerational Attenuation of Opioid Self-Administration as a Consequence of Adolescent Morphine Exposure. Neuropharmacology 113 (Pt A), 271–280. doi:10.1016/j.neuropharm.2016.10.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Voss, A. K., and Thomas, T. (2018). Histone Lysine and Genomic Targets of Histone Acetyltransferases in Mammals. Bioessays 40 (10), e1800078. doi:10.1002/bies.201800078

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, F., Xu, H., Zhao, H., Gelernter, J., and Zhang, H. (2016). DNA Co-methylation Modules in Postmortem Prefrontal Cortex Tissues of European Australians with Alcohol Use Disorders. Sci Rep 6, 19430. doi:10.1038/srep19430

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S. (2019). Historical Review: Opiate Addiction and Opioid Receptors. Cell Transplant 28 (3), 233–238. doi:10.1177/0963689718811060

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, J., Wang, T., Su, Z., Zhou, X., Xiang, Y., He, L., et al. (2018). Opioid Exposure Is Associated with Aberrant DNA Methylation of OPRM1 Promoter Region in a Chinese Han Population. Biochem Genet 56 (5), 451–458. doi:10.1007/s10528-018-9852-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Zadina, J. E., Hackler, L., Ge, L. J., and Kastin, A. J. (1997). A Potent and Selective Endogenous Agonist for the Mu-Opiate Receptor. Nature 386 (6624), 499–502. doi:10.1038/386499a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Wang, D., Johnson, A. D., Papp, A. C., and Sadée, W. (2005). Allelic Expression Imbalance of Human Mu Opioid Receptor (OPRM1) Caused by Variant A118G. J Biol Chem 280 (38), 32618–32624. doi:10.1074/jbc.M504942200

CrossRef Full Text | Google Scholar

Keywords: epigenetics, opioid use disorder (OUD), DNA methylation, mu opioid (MOP) receptor, African Americans

Citation: Blackwood CA and Cadet JL (2021) Epigenetic and Genetic Factors Associated With Opioid Use Disorder: Are These Relevant to African American Populations. Front. Pharmacol. 12:798362. doi: 10.3389/fphar.2021.798362

Received: 20 October 2021; Accepted: 07 December 2021;
Published: 22 December 2021.

Edited by:

Sandra Montagud Romero, University of Zaragoza, Spain

Reviewed by:

Sowmya Yelamanchili, University of Nebraska Medical Center, United States

Copyright © 2021 Blackwood and Cadet. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jean Lud Cadet, jcadet@intra.nida.nih.gov; Christopher A. Blackwood, Christopher.blackwood@nih.gov

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.