Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.997
Filter
1.
Commun Biol ; 7(1): 933, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095612

ABSTRACT

Desensitisation of the mu-opioid receptor (MOR) is proposed to underlie the initiation of opioid analgesic tolerance and previous work has shown that agonist-induced phosphorylation of the MOR C-tail contributes to this desensitisation. Moreover, phosphorylation is important for ß-arrestin recruitment to the receptor, and ligands of different efficacies induce distinct phosphorylation barcodes. The C-tail 370TREHPSTANT379 motif harbours Ser/Thr residues important for these regulatory functions. 375Ser is the primary phosphorylation site of a ligand-dependent, hierarchical, and sequential process, whereby flanking 370Thr, 376Thr and 379Thr get subsequently and rapidly phosphorylated. Here we used GRK KO cells, phosphosite specific antibodies and site-directed mutagenesis to evaluate the contribution of the different GRK subfamilies to ligand-induced phosphorylation barcodes and ß-arrestin2 recruitment. We show that both GRK2/3 and GRK5/6 subfamilies promote phosphorylation of 370Thr and 375Ser. Importantly, only GRK2/3 induce phosphorylation of 376Thr and 379Thr, and we identify these residues as key sites to promote robust ß-arrestin recruitment to the MOR. These data provide insight into the mechanisms of MOR regulation and suggest that the cellular complement of GRK subfamilies plays an important role in determining the tissue responses of opioid agonists.


Subject(s)
Receptors, Opioid, mu , beta-Arrestin 2 , Phosphorylation , beta-Arrestin 2/metabolism , beta-Arrestin 2/genetics , Humans , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/genetics , HEK293 Cells , Protein Binding , Animals , G-Protein-Coupled Receptor Kinases/metabolism , G-Protein-Coupled Receptor Kinases/genetics
2.
Clin Transl Sci ; 17(8): e13888, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39118392

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 ß-arrestin association with MOR r generated data that tested this prediction. Consistent with this in silico prediction, we show changes in morphine-mediated ß-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.


Subject(s)
Molecular Docking Simulation , Morphine , Polymorphism, Single Nucleotide , Receptors, Opioid, mu , beta-Arrestins , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Humans , beta-Arrestins/metabolism , Morphine/pharmacology , HEK293 Cells , Analgesics, Opioid/pharmacology , Analgesics, Opioid/metabolism , Protein Binding
3.
Addict Biol ; 29(7): e13422, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38949208

ABSTRACT

Opioid use disorder (OUD) is a multifaceted condition influenced by sex, genetic and environmental factors that could be linked with epigenetic changes. Understanding how these factors interact is crucial to understand and address the development and progression of this disorder. Our aim was to elucidate different potential epigenetic and genetic mechanisms between women and men that correlate with OUD under real-world pain unit conditions. Associations between analgesic response and the DNA methylation level of the opioid mu receptor (OPRM1) gene (CpG sites 1-5 selected in the promoter region) were evaluated in 345 long opioid-treated chronic non cancer pain: cases with OUD (n = 67) and controls (without OUD, n = 278). Cases showed younger ages, low employment status and quality of life, but higher morphine equivalent daily dose and psychotropic use, compared to the controls. The patients with OUD showed a significant decrease in OPRM1 DNA methylation, which correlated with clinical outcomes like pain relief, depression and different adverse events. Significant differences were found at the five CpG sites studied for men, and exclusively in women for CpG site 3, in relation to OUD diagnosis. These findings support the importance of epigenetics and sex as biological variables to be considered toward efficient OUD understanding and therapy development.


Subject(s)
Chronic Pain , DNA Methylation , Opioid-Related Disorders , Receptors, Opioid, mu , Adult , Female , Humans , Male , Middle Aged , Analgesics, Opioid/therapeutic use , Case-Control Studies , Chronic Pain/drug therapy , Chronic Pain/epidemiology , Chronic Pain/genetics , CpG Islands/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Opioid-Related Disorders/genetics , Quality of Life , Receptors, Opioid, mu/genetics , Sex Factors
4.
Clin Toxicol (Phila) ; 62(6): 357-363, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38946467

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.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, mu , Seizures , Tramadol , Humans , Tramadol/poisoning , Cross-Sectional Studies , Receptors, Opioid, mu/genetics , Male , Female , Adult , Iran , Analgesics, Opioid/poisoning , Analgesics, Opioid/adverse effects , Middle Aged , Seizures/genetics , Seizures/chemically induced , Young Adult , Polymorphism, Single Nucleotide , Drug Overdose/genetics , Genotype , Nausea/chemically induced , Nausea/genetics , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/genetics , Vomiting/chemically induced , Vomiting/genetics , Adolescent , Dizziness/chemically induced , Dizziness/genetics
5.
Neurosci Lett ; 837: 137898, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39013536

ABSTRACT

PURPOSE: Sex differences play a crucial role in understanding vulnerability to opioid addiction, yet there have been limited preclinical investigations of this effect during the transition from adolescence to adulthood. The present study compared the behaviors of male and female rodents in response to fentanyl treatment and targeted molecular correlates in the striatum and medial prefrontal cortex. MATERIALS AND METHODS: Thirty adolescent C57BL/6J mice underwent a 1-week fentanyl treatment with an escalating dose. In addition to evaluating locomotor activity and anxiety-related parameters, we also assessed naloxone-induced fentanyl acute withdrawal jumps. We employed real-time quantitative PCR (qPCR) to assess overall gene expression of dopaminergic receptors (Drd1, Drd2, Drd4 and Drd5) and the µ-opioid receptor Oprm1. The levels of epigenetic base modifications including 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) were assessed on CpG islands of relevant genes. RESULTS: Females had higher locomotor activity than males after chronic fentanyl treatment, and they exhibited higher fentanyl withdrawal jumping behavior induced by naloxone. Females also presented lower Drd4 gene expression and DNA methylation (5mC + 5hmC) in the striatum. We found that locomotor activity and fentanyl withdrawal jumps were negatively correlated with Drd4 methylation and gene expression in the striatum, respectively. CONCLUSIONS: The findings suggested that female mice displayed heightened sensitivity to the effects of fentanyl treatment during the transition from adolescence to adulthood. This effect may be associated with molecular alterations related to the Drd4 gene.


Subject(s)
Fentanyl , Mice, Inbred C57BL , Receptors, Opioid, mu , Sex Characteristics , Animals , Fentanyl/pharmacology , Male , Female , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Mice , DNA Methylation/drug effects , Analgesics, Opioid/pharmacology , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Locomotion/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, Dopamine/genetics , Receptors, Dopamine/metabolism , Naloxone/pharmacology , Behavior, Animal/drug effects , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/metabolism , Epigenesis, Genetic/drug effects
6.
Genes Brain Behav ; 23(3): e12906, 2024 06.
Article in English | MEDLINE | ID: mdl-38861664

ABSTRACT

Motherhood is a costly life-history transition accompanied by behavioral and neural plasticity necessary for offspring care. Motherhood in the monogamous prairie vole is associated with decreased pair bond strength, suggesting a trade-off between parental investment and pair bond maintenance. Neural mechanisms governing pair bonds and maternal bonds overlap, creating possible competition between the two. We measured mRNA expression of genes encoding receptors for oxytocin (oxtr), dopamine (d1r and d2r), mu-opioids (oprm1a), and kappa-opioids (oprk1a) within three brain areas processing salience of sociosensory cues (anterior cingulate cortex; ACC), pair bonding (nucleus accumbens; NAc), and maternal care (medial preoptic area; MPOA). We compared gene expression differences between pair bonded prairie voles that were never pregnant, pregnant (~day 16 of pregnancy), and recent mothers (day 3 of lactation). We found greater gene expression in the NAc (oxtr, d2r, oprm1a, and oprk1a) and MPOA (oxtr, d1r, d2r, oprm1a, and oprk1a) following the transition to motherhood. Expression for all five genes in the ACC was greatest for females that had been bonded for longer. Gene expression within each region was highly correlated, indicating that oxytocin, dopamine, and opioids comprise a complimentary gene network for social signaling. ACC-NAc gene expression correlations indicated that being a mother (oxtr and d1r) or maintaining long-term pair bonds (oprm1a) relies on the coordination of different signaling systems within the same circuit. Our study suggests the maternal brain undergoes changes that prepare females to face the trade-off associated with increased emotional investment in offspring, while also maintaining a pair bond.


Subject(s)
Arvicolinae , Maternal Behavior , Nucleus Accumbens , Pair Bond , Receptors, Opioid, mu , Animals , Female , Arvicolinae/genetics , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Maternal Behavior/physiology , Nucleus Accumbens/metabolism , Pregnancy , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , Gyrus Cinguli/metabolism , Preoptic Area/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism
7.
Science ; 384(6700): eadn0886, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843332

ABSTRACT

In addition to their intrinsic rewarding properties, opioids can also evoke aversive reactions that protect against misuse. Cellular mechanisms that govern the interplay between opioid reward and aversion are poorly understood. We used whole-brain activity mapping in mice to show that neurons in the dorsal peduncular nucleus (DPn) are highly responsive to the opioid oxycodone. Connectomic profiling revealed that DPn neurons innervate the parabrachial nucleus (PBn). Spatial and single-nuclei transcriptomics resolved a population of PBn-projecting pyramidal neurons in the DPn that express µ-opioid receptors (µORs). Disrupting µOR signaling in the DPn switched oxycodone from rewarding to aversive and exacerbated the severity of opioid withdrawal. These findings identify the DPn as a key substrate for the abuse liability of opioids.


Subject(s)
Analgesics, Opioid , Avoidance Learning , Opioid-Related Disorders , Oxycodone , Parabrachial Nucleus , Prefrontal Cortex , Receptors, Opioid, mu , Reward , Animals , Male , Mice , Analgesics, Opioid/pharmacology , Connectome , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology , Opioid-Related Disorders/metabolism , Oxycodone/pharmacology , Parabrachial Nucleus/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Pyramidal Cells/metabolism , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/genetics , Substance Withdrawal Syndrome/metabolism , Transcriptome
8.
Sci Rep ; 14(1): 11273, 2024 05 17.
Article in English | MEDLINE | ID: mdl-38760456

ABSTRACT

To investigate the association between three selected pain polymorphisms and clinical, functional, sensory-related, psychophysical, psychological or cognitive variables in a sample of women with fibromyalgia (FMS). One hundred twenty-three (n = 123) women with FMS completed demographic (age, height, weight), clinical (years with pain, intensity of pain at rest and during daily living activities), functional (quality of life, physical function), sensory-related (sensitization-associated and neuropathic-associated symptoms), psychophysical (pressure pain thresholds), psychological (sleep quality, depressive and anxiety level) and cognitive (pain catastrophizing, kinesiophobia) variables. Those three genotypes of the OPRM1 rs1799971, HTR1B rs6296 and COMT rs4680 single nucleotide polymorphisms were obtained by polymerase chain reactions from no-stimulated whole saliva collection. No significant differences in demographic, clinical, functional, sensory-related, psychophysical, psychological and cognitive variables according to OPRM1 rs1799971, HTR1B rs6296 or COMT rs4680 genotype were identified in our sample of women with FMS. A multilevel analysis did not either reveal any significant gene-to-gene interaction between OPRM1 rs1799971 x HTR1B rs6296, OPRM1 rs1799971 x COMT rs4680 and HTR1B rs6296 x COMT rs4680 for any of the investigated outcomes. This study revealed that three single nucleotide polymorphisms, OPRM1 rs1799971, HTR1B rs6296 or COMT rs4680, mostly associated with chronic pain were not involved in phenotyping features of FMS. Potential gene-to-gene interaction and their association with clinical phenotype in women with FMS should be further investigated in future studies including large sample sizes.


Subject(s)
Catechol O-Methyltransferase , Fibromyalgia , Polymorphism, Single Nucleotide , Receptor, Serotonin, 5-HT1B , Receptors, Opioid, mu , Humans , Fibromyalgia/genetics , Female , Catechol O-Methyltransferase/genetics , Receptors, Opioid, mu/genetics , Middle Aged , Adult , Receptor, Serotonin, 5-HT1B/genetics , Phenotype , Genotype , Genetic Predisposition to Disease , Quality of Life
9.
Neuropharmacology ; 255: 110019, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38810926

ABSTRACT

The endogenous opioid system has been implicated in alcohol consumption and preference in both humans and animals. The mu opioid receptor (MOR) is expressed on multiple cells in the striatum, however little is known about the contributions of specific MOR populations to alcohol drinking behaviors. The current study used mice with a genetic deletion of MOR in cholinergic cells (ChAT-Cre/Oprm1fl/fl) to examine the role of MORs expressed in cholinergic interneurons (CINs) in home cage self-administration paradigms. Male and female ChAT-Cre/Oprm1fl/fl mice were generated and heterozygous Cre+ (knockout) and Cre- (control) mice were tested for alcohol consumption in two drinking paradigms: limited access "Drinking in the Dark" and intermittent access. Quinine was added to the drinking bottles in the DID experiment to test aversion-resistant, "compulsive" drinking. Nicotine and sucrose drinking were also assessed so comparisons could be made with other rewarding substances. Cholinergic MOR deletion did not influence consumption or preference for ethanol (EtOH) in either drinking task. Differences were observed in aversion-resistance in males with Cre + mice tolerating lower concentrations of quinine than Cre-. In contrast to EtOH, preference for nicotine was reduced following cholinergic MOR deletion while sucrose consumption and preference was increased in Cre+ (vs. Cre-) females. Locomotor activity was also greater in females following the deletion. These results suggest that cholinergic MORs participate in preference for rewarding substances. Further, while they are not required for consumption of alcohol alone, cholinergic MORs may influence the tendency to drink despite negative consequences.


Subject(s)
Alcohol Drinking , Mice, Knockout , Quinine , Receptors, Opioid, mu , Reward , Animals , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Male , Female , Mice , Quinine/pharmacology , Quinine/administration & dosage , Alcohol Drinking/genetics , Alcohol Drinking/psychology , Nicotine/pharmacology , Ethanol/pharmacology , Ethanol/administration & dosage , Cholinergic Neurons/drug effects , Cholinergic Neurons/physiology , Cholinergic Neurons/metabolism , Self Administration , Sucrose/administration & dosage , Avoidance Learning/drug effects , Avoidance Learning/physiology , Interneurons/drug effects , Interneurons/physiology , Interneurons/metabolism
10.
Nat Commun ; 15(1): 3544, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740791

ABSTRACT

G-protein-coupled receptors (GPCRs) play pivotal roles in various physiological processes. These receptors are activated to different extents by diverse orthosteric ligands and allosteric modulators. However, the mechanisms underlying these variations in signaling activity by allosteric modulators remain largely elusive. Here, we determine the three-dimensional structure of the µ-opioid receptor (MOR), a class A GPCR, in complex with the Gi protein and an allosteric modulator, BMS-986122, using cryogenic electron microscopy. Our results reveal that BMS-986122 binding induces changes in the map densities corresponding to R1673.50 and Y2545.58, key residues in the structural motifs conserved among class A GPCRs. Nuclear magnetic resonance analyses of MOR in the absence of the Gi protein reveal that BMS-986122 binding enhances the formation of the interaction between R1673.50 and Y2545.58, thus stabilizing the fully-activated conformation, where the intracellular half of TM6 is outward-shifted to allow for interaction with the Gi protein. These findings illuminate that allosteric modulators like BMS-986122 can potentiate receptor activation through alterations in the conformational dynamics in the core region of GPCRs. Together, our results demonstrate the regulatory mechanisms of GPCRs, providing insights into the rational development of therapeutics targeting GPCRs.


Subject(s)
Cryoelectron Microscopy , Receptors, Opioid, mu , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/genetics , Allosteric Regulation , Humans , Protein Binding , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , HEK293 Cells , Ligands , Models, Molecular , Protein Conformation
11.
J Oral Rehabil ; 51(8): 1542-1554, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38725226

ABSTRACT

BACKGROUND: Single nucleotide polymorphisms (SNPs) may influence pain susceptibility and impact treatment response in pain-related temporomandibular disorders (TMDp). OBJECTIVE: Explore the role of COMT (rs4646310, rs6269, rs4818, rs4680) and OPRM1 (rs1799971) genotypes in regulating treatment response. METHODS: Sixty TMDp patients (55 females and 5 males), diagnosed with the Diagnostic Criteria for TMD (DC/TMD), underwent standardised treatment (information and education, home physical therapy, occlusal splint) for 6 months. Treatment outcomes included: pain intensity, pain-free mouth opening, jaw functional limitation, depression, and anxiety. Genotyping for COMT and OPRM1 SNPs was performed using DNA from buccal mucosa swabs and TaqMan assays. Statistical analysis was carried out to compare the changes in treatment outcomes and the influence of genotypes on treatment response. RESULTS: Significantly less pain reduction was observed in minor allele carriers of rs4646310, and rs4680 compared to dominant homozygous (p < .025). Minor allele carriers of rs1799971 and rs4646310 demonstrated worsening in pain-free mouth opening while dominant homozygous exhibited improvement (p < .025). Significantly less anxiety reduction was observed in minor allele carriers of rs4646310 compared to dominant homozygous (p = .003). Of the all variables assessed in the regression model, carrying a minor allele of rs1799971 predicted a poorer treatment response considering pain-free mouth opening while carrying a minor allele of rs4646310 predicted less pain and less anxiety reduction. CONCLUSION: Our findings indicate that certain SNP variants of the COMT and OPRM1 genes were associated with poorer treatment response and may therefore play a significant role in the classification of TMDp patients. Also, assessment of patient genotype could potentially aid in predicting treatment response.


Subject(s)
Catechol O-Methyltransferase , Facial Pain , Genotype , Pain Measurement , Polymorphism, Single Nucleotide , Receptors, Opioid, mu , Temporomandibular Joint Disorders , Humans , Female , Male , Temporomandibular Joint Disorders/genetics , Temporomandibular Joint Disorders/therapy , Temporomandibular Joint Disorders/physiopathology , Adult , Treatment Outcome , Catechol O-Methyltransferase/genetics , Facial Pain/genetics , Facial Pain/therapy , Facial Pain/physiopathology , Receptors, Opioid, mu/genetics , Middle Aged , Occlusal Splints , Young Adult , Genetic Predisposition to Disease , Alleles
12.
Am J Psychiatry ; 181(5): 403-411, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38706338

ABSTRACT

OBJECTIVE: There have been no well-controlled and well-powered comparative trials of topiramate with other pharmacotherapies for alcohol use disorder (AUD), such as naltrexone. Moreover, the literature is mixed on the effects of two polymorphisms-rs2832407 (in GRIK1) and rs1799971 (in OPRM1)-on response to topiramate and naltrexone, respectively. The authors sought to examine the comparative effectiveness of topiramate and naltrexone in improving outcomes in AUD and to examine the role of the rs2832407 and rs1799971 polymorphisms, respectively, on response to these medications. METHODS: In a 12-week, double-blind, placebo-controlled, randomized, multisite, genotype-stratified (rs2832407 and rs1799971) clinical trial comparing topiramate and naltrexone in treating AUD, 147 patients with AUD were randomly assigned to treatment with topiramate or naltrexone, stratified by genotype (rs2832407*CC and *AC/AA genotypes and rs1799971*AA and *AG/GG genotypes). The predefined primary outcome was number of heavy drinking days per week. Predefined secondary outcomes included standard drinks per drinking day per week, body mass index (BMI), craving, markers of liver injury, mood, and adverse events. RESULTS: For the number of heavy drinking days per week, there was a near-significant time-by-treatment interaction. For the number of standard drinks per drinking day per week, there was a significant time-by-treatment interaction, which favored topiramate. There were significant time-by-treatment effects, with greater reductions observed with topiramate than naltrexone for BMI, craving, and gamma-glutamyltransferase level. Withdrawal due to side effects occurred in 8% and 5% of the topiramate and naltrexone groups, respectively. Neither polymorphism showed an effect on treatment response. CONCLUSIONS: Topiramate is at least as effective and safe as the first-line medication, naltrexone, in reducing heavy alcohol consumption, and superior in reducing some clinical outcomes. Neither rs2832407 nor rs1799971 had effects on topiramate and naltrexone treatments, respectively.


Subject(s)
Alcoholism , Genotype , Naltrexone , Receptors, Kainic Acid , Topiramate , Humans , Topiramate/therapeutic use , Naltrexone/therapeutic use , Double-Blind Method , Male , Female , Alcoholism/drug therapy , Alcoholism/genetics , Adult , Middle Aged , Receptors, Kainic Acid/genetics , Receptors, Opioid, mu/genetics , Treatment Outcome , Narcotic Antagonists/therapeutic use , Polymorphism, Single Nucleotide , Craving/drug effects , Fructose/analogs & derivatives , Fructose/therapeutic use
13.
Neurosci Lett ; 834: 137846, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38821204

ABSTRACT

OPRM1 gene encoding mu-opioid receptor (MOR) is the primary candidate gene for buprenorphine (BUP) pharmacogenetics. OPRM1 undergoes alternative splicing leading to multiple MOR subtypes. Thus, in the current study 2 SNPs (rs1799972 and rs562859) were selected due to evidence for their contribution to alternative splicing of OPRM1. The effects of 2 SNPs of OPRM1 gene on plasma buprenorphine and norbuprenorphine levels in a sample of 233 OUD patients receiving BUP/naloxone were examined. Polymorphisms were analyzed by PCR and RFLP. BUP and norbuprenorphine concentrations in plasma were measured by LC-MS/MS. OPRM1 rs2075572 GC + CC (0.12 ng/ml) had significantly higher plasma BUP level compared to GG (0.084 ng/ml) (p = 0.043). Although there was not a statistically significant difference between OPRM1 rs562859 genotypes (p = 0.46), patients with OPRM1 rs562859 CT + TT had higher plasma BUP and BUP-related values as compared to those with CC. In conclusion, the effect of OPRM1 rs2075572 on BUP levels in opioid users' plasma was shown in a Caucasian population for the first time. On the other hand, OPRM1 rs562859 seems not to influence the BUP pharmacology.


Subject(s)
Buprenorphine , Opioid-Related Disorders , Polymorphism, Single Nucleotide , Receptors, Opioid, mu , Humans , Receptors, Opioid, mu/genetics , Male , Female , Adult , Buprenorphine/blood , Buprenorphine/therapeutic use , Buprenorphine/analogs & derivatives , Polymorphism, Single Nucleotide/genetics , Opioid-Related Disorders/genetics , Opioid-Related Disorders/blood , Middle Aged , Analgesics, Opioid/blood , Genotype
14.
J Neurophysiol ; 132(1): 108-129, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38748514

ABSTRACT

µ-Opioid receptors (MORs) are responsible for mediating both the analgesic and respiratory effects of opioid drugs. By binding to MORs in brainstem regions involved in controlling breathing, opioids produce respiratory depressive effects characterized by slow and shallow breathing, with potential cardiorespiratory arrest and death during overdose. To better understand the mechanisms underlying opioid-induced respiratory depression, thorough knowledge of the regions and cellular subpopulations that may be vulnerable to modulation by opioid drugs is needed. Using in situ hybridization, we determined the distribution and coexpression of Oprm1 (gene encoding MORs) mRNA with glutamatergic (Vglut2) and neurokinin-1 receptor (Tacr1) mRNA in medullary and pontine regions involved in breathing control and modulation. We found that >50% of cells expressed Oprm1 mRNA in the preBötzinger complex (preBötC), nucleus tractus solitarius (NTS), nucleus ambiguus (NA), postinspiratory complex (PiCo), locus coeruleus (LC), Kölliker-Fuse nucleus (KF), and the lateral and medial parabrachial nuclei (LBPN and MPBN, respectively). Among Tacr1 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, Bötzinger complex (BötC), PiCo, LC, raphe magnus nucleus, KF, LPBN, and MPBN, whereas among Vglut2 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, BötC, PiCo, LC, KF, LPBN, and MPBN. Taken together, our study provides a comprehensive map of the distribution and coexpression of Oprm1, Tacr1, and Vglut2 mRNA in brainstem regions that control and modulate breathing and identifies Tacr1 and Vglut2 mRNA-expressing cells as subpopulations with potential vulnerability to modulation by opioid drugs.NEW & NOTEWORTHY Opioid drugs can cause serious respiratory side-effects by binding to µ-opioid receptors (MORs) in brainstem regions that control breathing. To better understand the regions and their cellular subpopulations that may be vulnerable to modulation by opioids, we provide a comprehensive map of Oprm1 (gene encoding MORs) mRNA expression throughout brainstem regions that control and modulate breathing. Notably, we identify glutamatergic and neurokinin-1 receptor-expressing cells as potentially vulnerable to modulation by opioid drugs and worthy of further investigation using targeted approaches.


Subject(s)
Receptors, Neurokinin-1 , Receptors, Opioid, mu , Vesicular Glutamate Transport Protein 2 , Animals , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/genetics , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-1/genetics , Mice , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Male , Brain Stem/metabolism , Brain Stem/drug effects , Mice, Inbred C57BL , RNA, Messenger/metabolism , RNA, Messenger/genetics , Respiratory Center/metabolism , Respiratory Center/drug effects
15.
Transl Psychiatry ; 14(1): 181, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38580654

ABSTRACT

The endogenous opioid system is thought to play an important role in mother-infant attachment. In infant rhesus macaques, variation in the µ-opioid receptor gene (OPRM1) is related to differences in attachment behavior that emerges following repeated separation from the mother; specifically, infants carrying at least one copy of the minor G allele of the OPRM1 C77G polymorphism show heightened and more persistent separation distress, as well as a pattern of increased contact-seeking behavior directed towards the mother during reunions (at the expense of affiliation with other group members). Research in adult humans has also linked the minor G allele of the analogous OPRM1 A118G polymorphism with greater interpersonal sensitivity. Adopting an interactionist approach, we examined whether OPRM1 A118G genotype and maternal (in)sensitivity are associated with child attachment style, predicting that children carrying the G allele may be more likely to develop an ambivalent attachment pattern in response to less sensitive maternal care. The sample consisted of 191 mothers participating with their children (n = 223) in the Maternal Adversity, Vulnerability and Neurodevelopment (MAVAN) project, a community-based, birth cohort study of Canadian mothers and their children assessed longitudinally across the child's development. Maternal sensitivity was coded from at-home mother-child interactions videotaped when the child was 18 months of age. Child attachment was assessed at 36 months using the Strange Situation paradigm. As predicted, G allele carriers, but not AA homozygotes, showed increasing odds of being classified as ambivalently attached with decreasing levels of maternal sensitivity. Paralleling earlier non-human animal research, this work provides support for the theory that endogenous opioids contribute to the expression of attachment behaviors in humans.


Subject(s)
Mother-Child Relations , Polymorphism, Genetic , Adult , Female , Humans , Canada , Cohort Studies , Genotype , Polymorphism, Single Nucleotide , Receptors, Opioid, mu/genetics
16.
Minerva Anestesiol ; 90(5): 386-396, 2024 05.
Article in English | MEDLINE | ID: mdl-38619184

ABSTRACT

BACKGROUND: Opioids are widely used in chronic non-cancer pain (CNCP) management. However, they remain controversial due to serious risk of causing opioid use disorder (OUD). Our main aim was to develop a predictive model for future clinical translation that include pharmacogenetic markers. METHODS: An observational study was conducted in 806 pre-screened Spanish CNCP patients, under long-term use of opioids, to compare cases (with OUD, N.=137) with controls (without OUD, N.=669). Mu-opioid receptor 1 (OPRM1, A118G, rs1799971) and catechol-O-methyltransferase (COMT, G472A, rs4680) genetic variants plus cytochrome P450 2D6 (CYP2D6) liver enzyme phenotypes were analyzed. Socio-demographic, clinical and pharmacological outcomes were also registered. A logistic regression model was performed. The model performance and diagnostic accuracy were calculated. RESULTS: OPRM1-AA genotype and CYP2D6 poor and ultrarapid metabolizers together with three other potential predictors: 1) age; 2) work disability; 3) oral morphine equivalent daily dose (MEDD), were selected with a satisfactory diagnostic accuracy (sensitivity: 0.82 and specificity: 0.85), goodness of fit (P=0.87) and discrimination (0.89). Cases were ten-year younger with lower incomes, more sleep disturbances, benzodiazepines use, and history of substance use disorder in front of controls. CONCLUSIONS: Functional polymorphisms related to OPRM1 variant and CYP2D6 phenotypes may predict a higher OUD risk. Established risk factors such as young age, elevated MEDD and lower incomes were identified. A predictive model is expected to be implemented in clinical setting among CNCP patients under long-term opioids use.


Subject(s)
Chronic Pain , Opioid-Related Disorders , Humans , Male , Female , Chronic Pain/drug therapy , Chronic Pain/genetics , Middle Aged , Opioid-Related Disorders/genetics , Adult , Retrospective Studies , Cohort Studies , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/adverse effects , Pharmacogenetics , Receptors, Opioid, mu/genetics , Cytochrome P-450 CYP2D6/genetics , Catechol O-Methyltransferase/genetics , Aged , Genotype
17.
Psychopharmacology (Berl) ; 241(7): 1435-1446, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38503843

ABSTRACT

RATIONALE: Transgenerational effects of preconception morphine exposure in female rats have been reported which suggest that epigenetic modifications triggered by female opioid exposure, even when that exposure ends several weeks prior to pregnancy, has significant ramifications for their future offspring. OBJECTIVE: The current study compares two mouse strains with well-established genetic variation in their response to mu opioid receptor agonists, C57BL/6J (BL6) and 129S1/svlmJ (129) to determine whether genetic background modifies the impact of preconception opioid exposure. METHODS: Adolescent females from both strains were injected daily with morphine for a total of 10 days using an increasing dosing regimen with controls receiving saline. Several weeks after their final injection, aged-matched BL6 and 129 morphine (Mor-F0) or saline (Sal-F0) females were mated with drug naïve males to generate Mor-F1 and Sal-F1 offspring, respectively. As adults, F1 mice were made morphine dependent using thrice daily morphine injections for 4 days. On day 5, mice were administered either saline or morphine followed 3 h later by naloxone. Behavioral and physiological signs of withdrawal were then measured. RESULTS: Regardless of strain or sex, morphine-dependent Mor-F1 mice had significantly lower levels of withdrawal-induced corticosterone but significantly higher glucose levels when compared to Sal-F1 controls. In contrast, both strain- and preconception opioid exposure effects on physical signs of morphine dependence were observed.


Subject(s)
Analgesics, Opioid , Mice, Inbred C57BL , Morphine Dependence , Morphine , Receptors, Opioid, mu , Substance Withdrawal Syndrome , Animals , Female , Morphine/pharmacology , Morphine/administration & dosage , Substance Withdrawal Syndrome/metabolism , Mice , Male , Morphine Dependence/metabolism , Pregnancy , Analgesics, Opioid/pharmacology , Analgesics, Opioid/administration & dosage , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/genetics , Mice, 129 Strain , Naloxone/pharmacology , Naloxone/administration & dosage , Species Specificity , Narcotic Antagonists/pharmacology , Narcotic Antagonists/administration & dosage , Corticosterone/blood , Prenatal Exposure Delayed Effects/metabolism
18.
Int J Mol Sci ; 25(5)2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38474311

ABSTRACT

The main aims of the present study were to explore the relationship of the OPRM1 gene rs1074287 polymorphism in alcohol-dependent women with their personality traits and to try to find out whether any specific features may influence alcohol cravings and be a prognostic for alcohol dependency and treatment in AUD women. Our study found a notable correlation between openness and the interaction of the ORIM1 gene and AUD. The alcohol use disorder subjects with genotype AG showed a higher level of openness compared to the control group with genotypes AG (p = 0.0001) and AA (p = 0.0125). The alcohol use disorder subjects with the AA genotype displayed higher levels of openness than the control group with genotype AG (p = 0.0271). However, the alcohol use disorder subjects with the AA genotype displayed lower levels of openness than the control group with genotype GG (p = 0.0212). Our study indicates that openness as a personality trait is correlated with the OPRM1 gene rs1074287 polymorphism in alcohol-dependent women. These are the first data and results exploring such a relationship between opioid and alcohol pathways and the mental construction of AUD women. Personality traits such as openness to experience and neuroticism might play major roles in the addiction mechanism, especially in genetically predisposed females, independent of the reward system involved in the emotional disturbances that coexist with anxiety and depression.


Subject(s)
Alcoholism , Genetic Predisposition to Disease , Personality , Receptors, Opioid, mu , Female , Humans , Alcoholism/genetics , Alcoholism/psychology , Ethanol , Genotype , Polymorphism, Single Nucleotide , Receptors, Opioid, mu/genetics
19.
Psychiatry Clin Neurosci ; 78(5): 300-308, 2024 May.
Article in English | MEDLINE | ID: mdl-38403942

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.


Subject(s)
Catechol O-Methyltransferase , Pain , Polymorphism, Single Nucleotide , Receptors, Opioid, mu , Humans , Catechol O-Methyltransferase/genetics , Receptors, Opioid, mu/genetics , Male , Adult , Female , Young Adult , Pain/genetics , Pain/physiopathology , Magnetic Resonance Imaging , Pain Perception/physiology , Brain/physiopathology , Functional Neuroimaging
20.
Sci Rep ; 14(1): 3411, 2024 02 10.
Article in English | MEDLINE | ID: mdl-38341456

ABSTRACT

Cancer pain is the most feared symptom at end of life. Methadone has advantages over other opioids but is associated with significant variability in clinical response, making dosing challenging in practice. OPRM1 is the most studied pharmacogene associated with the pharmacodynamics of opioids, however reports on the association of the A118G polymorphism on opioid dose requirements are conflicting, with no reports including methadone as the primary intervention. This association study on OPRM1 A118G and response to methadone for pain management, includes a review of this genetic factor's role in inter-patient variability. Fifty-four adult patients with advanced cancer were recruited in a prospective, multi-centre, open label dose individualization study. Patient characteristics were not shown to influence methadone response, and no significant associations were observed for methadone dose or pain score. The findings of our review of association studies for OPRM1 A118G in advanced cancer pain demonstrate the importance of taking ancestry into account. While our sample size was small, our results were consistent with European populations, but in contrast to studies in Chinese patients, where carriers of the A118G polymorphism were associated with higher opioid dose requirements. Pharmacogenetic studies in palliative care are challenging, continued contribution will support future genotype-based drug dosing guidelines.


Subject(s)
Cancer Pain , Neoplasms , Adult , Humans , Analgesics, Opioid/therapeutic use , Cancer Pain/drug therapy , Cancer Pain/genetics , Genotype , Methadone/therapeutic use , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/genetics , Pain Management , Polymorphism, Single Nucleotide , Prospective Studies , Receptors, Opioid, mu/genetics
SELECTION OF CITATIONS
SEARCH DETAIL