Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Addict Biol ; 29(7): e13422, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38949208

RESUMO

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.


Assuntos
Dor Crônica , Metilação de DNA , Epigênese Genética , Transtornos Relacionados ao Uso de Opioides , Receptores Opioides mu , Humanos , Receptores Opioides mu/genética , Metilação de DNA/genética , Masculino , Feminino , Dor Crônica/genética , Dor Crônica/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/genética , Pessoa de Meia-Idade , Adulto , Fatores Sexuais , Analgésicos Opioides/uso terapêutico , Estudos de Casos e Controles , Ilhas de CpG/genética , Qualidade de Vida
2.
Minerva Anestesiol ; 90(5): 386-396, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38619184

RESUMO

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.


Assuntos
Dor Crônica , Transtornos Relacionados ao Uso de Opioides , Humanos , Masculino , Feminino , Dor Crônica/tratamento farmacológico , Dor Crônica/genética , Pessoa de Meia-Idade , Transtornos Relacionados ao Uso de Opioides/genética , Adulto , Estudos Retrospectivos , Estudos de Coortes , Analgésicos Opioides/uso terapêutico , Analgésicos Opioides/efeitos adversos , Farmacogenética , Receptores Opioides mu/genética , Citocromo P-450 CYP2D6/genética , Catecol O-Metiltransferase/genética , Idoso , Genótipo
3.
Addict Biol ; 27(1): e13099, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34611967

RESUMO

Polygenic risk scores (PRS) represent an individual's summed genetic risk for a trait and can serve as biomarkers for disease. Less is known about the utility of PRS as a means to quantify genetic risk for substance use disorders (SUDs) than for many other traits. Nonetheless, the growth of large, electronic health record-based biobanks makes it possible to evaluate the association of SUD PRS with other traits. We calculated PRS for smoking initiation, alcohol use disorder (AUD), and opioid use disorder (OUD) using summary statistics from the Million Veteran Program sample. We then tested the association of each PRS with its primary phenotype in the Penn Medicine BioBank (PMBB) using all available genotyped participants of African or European ancestry (AFR and EUR, respectively) (N = 18,612). Finally, we conducted phenome-wide association analyses (PheWAS) separately by ancestry and sex to test for associations across disease categories. Tobacco use disorder was the most common SUD in the PMBB, followed by AUD and OUD, consistent with the population prevalence of these disorders. All PRS were associated with their primary phenotype in both ancestry groups. PheWAS results yielded cross-trait associations across multiple domains, including psychiatric disorders and medical conditions. SUD PRS were associated with their primary phenotypes; however, they are not yet predictive enough to be useful diagnostically. The cross-trait associations of the SUD PRS are indicative of a broader genetic liability. Future work should extend findings to additional population groups and for other substances of abuse.


Assuntos
Comorbidade , Registros Eletrônicos de Saúde/estatística & dados numéricos , Predisposição Genética para Doença/genética , Transtornos Relacionados ao Uso de Substâncias/etnologia , Transtornos Relacionados ao Uso de Substâncias/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Alcoolismo/etnologia , Alcoolismo/genética , População Negra/genética , Feminino , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Herança Multifatorial , Transtornos Relacionados ao Uso de Opioides/etnologia , Transtornos Relacionados ao Uso de Opioides/genética , Fenótipo , Fatores de Risco , Fatores Sexuais , Tabagismo/etnologia , Tabagismo/genética , População Branca/genética
4.
Mol Psychiatry ; 27(3): 1742-1753, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34759357

RESUMO

Substance use disorders (SUDs) are moderately to highly heritable and are in part cross-transmitted genetically, as observed in twin and family studies. We performed exome-focused genotyping to examine the cross-transmission of four SUDs: alcohol use disorder (AUD, n = 4487); nicotine use disorder (NUD, n = 4394); cannabis use disorder (CUD, n = 954); and nonmedical prescription opioid use disorder (NMPOUD, n = 346) within a large nationally representative sample (n = 36,309), the National Epidemiologic Survey on Alcohol and Related Conditions-III (NESARC-III). All diagnoses were based on in-person structured psychiatric interview (AUDADIS-5). SUD cases were compared alone and together to 3959 "super controls" who had neither a SUD nor a psychiatric disorder using an exome-focused array assaying 363,496 SNPs, yielding a representative view of within-disorder and cross-disorder genetic influences on SUDs. The 29 top susceptibility genes for one or more SUDs overlapped highly with genes previously implicated by GWAS of SUD. Polygenic scores (PGS) were computed within the European ancestry (EA) component of the sample (n = 12,505) using summary statistics from each of four clinically distinct SUDs compared to the 3959 "super controls" but then used for two distinctly different purposes: to predict SUD severity (mild, moderate, or severe) and to predict each of the other 3 SUDs. Our findings based on PGS highlight shared and unshared genetic contributions to the pathogenesis of SUDs, confirming the strong cross-inheritance of AUD and NUD as well as the distinctiveness of inheritance of opioid use disorder.


Assuntos
Transtornos Relacionados ao Uso de Álcool , Alcoolismo , Transtornos Relacionados ao Uso de Opioides , Transtornos Relacionados ao Uso de Substâncias , Tabagismo , Consumo de Bebidas Alcoólicas , Alcoolismo/psicologia , Comorbidade , Humanos , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Transtornos Relacionados ao Uso de Opioides/genética , Transtornos Relacionados ao Uso de Substâncias/epidemiologia , Transtornos Relacionados ao Uso de Substâncias/genética , Transtornos Relacionados ao Uso de Substâncias/psicologia , Tabagismo/psicologia
5.
Drug Alcohol Depend ; 229(Pt B): 109115, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34710714

RESUMO

BACKGROUND: Machine learning (ML) models are beginning to proliferate in psychiatry, however machine learning models in psychiatric genetics have not always accounted for ancestry. Using an empirical example of a proposed genetic test for OUD, and exploring a similar test for tobacco dependence and a simulated binary phenotype, we show that genetic prediction using ML is vulnerable to ancestral confounding. METHODS: We utilize five ML algorithms trained with 16 brain reward-derived "candidate" SNPs proposed for commercial use and examine their ability to predict OUD vs. ancestry in an out-of-sample test set (N = 1000, stratified into equal groups of n = 250 cases and controls each of European and African ancestry). We rerun analyses with 8 random sets of allele-frequency matched SNPs. We contrast findings with 11 genome-wide significant variants for tobacco smoking. To document generalizability, we generate and test a random phenotype. RESULTS: None of the 5 ML algorithms predict OUD better than chance when ancestry was balanced but were confounded with ancestry in an out-of-sample test. In addition, the algorithms preferentially predicted admixed subpopulations. Random sets of variants matched to the candidate SNPs by allele frequency produced similar bias. Genome-wide significant tobacco smoking variants were also confounded by ancestry. Finally, random SNPs predicting a random simulated phenotype show that the bias attributable to ancestral confounding could impact any ML-based genetic prediction. CONCLUSIONS: Researchers and clinicians are encouraged to be skeptical of claims of high prediction accuracy from ML-derived genetic algorithms for polygenic traits like addiction, particularly when using candidate variants.


Assuntos
Herança Multifatorial , Transtornos Relacionados ao Uso de Opioides , População Negra/genética , Humanos , Aprendizado de Máquina , Transtornos Relacionados ao Uso de Opioides/genética , Polimorfismo de Nucleotídeo Único/genética
6.
Biochem Biophys Res Commun ; 558: 8-13, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33894675

RESUMO

Increasing evidences suggest the involvement of disrupted circadian clock in various pathologies including stroke and substance abuse. Here we took an attempt to do a comparative study on the regulation of circadian clock gene expression under two pathological circumstances - Opioid addiction and Ischemic stroke in the same cell line model (human neuroblastoma SH-SY5Y cells). To mimic in vivo ischemic stroke condition cells were placed in a hypoxia chamber and incubated for 10 h in balanced salt solution lacking glucose, aerated with an anaerobic gas mixture (95% N2 and 5% C02). For opioid addiction cells were treated with morphine sulphate at 10 µM dose for 48 h. We found that although circadian clock gets disturbed in both states, pattern of alteration of clock gene expressions were different and change was more severe in ischemic stroke than addiction. Interestingly, increase in expression of Cry1 showed as a common factor to both the diseases. This paper also emphasizes the interconnection between the severities of neuronal injury induced by ischemic stroke or opioid abuse to circadian system. Finally, this study will further enrich our knowledge towards the pattern of circadian rhythm disturbances under different pathological states.


Assuntos
Relógios Circadianos/genética , Relógios Circadianos/fisiologia , Criptocromos/genética , Criptocromos/fisiologia , AVC Isquêmico/genética , AVC Isquêmico/fisiopatologia , Transtornos Relacionados ao Uso de Opioides/genética , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Hipóxia Celular , Linhagem Celular , Sobrevivência Celular , Glucose/deficiência , Humanos , AVC Isquêmico/patologia , Modelos Biológicos , Morfina/administração & dosagem , Transtornos Relacionados ao Uso de Opioides/patologia , Regulação para Cima/efeitos dos fármacos
7.
Exp Neurol ; 341: 113710, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33781732

RESUMO

Chronic exposure to opioids typically results in adverse consequences. Opioid use disorder (OUD) is a disease of the CNS with behavioral, psychological, neurobiological, and medical manifestations. OUD induces a variety of changes of neurotransmitters/neuropeptides in the nervous system. Existing pharmacotherapy, such as opioid maintenance therapy (OMT) is the mainstay for the treatment of OUD, however, current opioid replacement therapy is far from effective for the majority of patients. Pharmacological therapy for OUD has been challenging for many reasons including debilitating side-effects. Therefore, developing an effective, non-pharmacological approach would be a critical advancement in improving and expanding treatment for OUD. Viral vector mediated gene therapy provides a potential new approach for treating opioid abused patients. Gene therapy can supply targeting gene products directly linked to the mechanisms of OUD to restore neurotransmitter and/or neuropeptides imbalance, and avoid the off-target effects of systemic administration of drugs. The most commonly used viral vectors in rodent studies of treatment of opioid-used disorder are based on recombinant adenovirus (AV), adeno-associated virus (AAV), lentiviral (LV) vectors, and herpes simplex virus (HSV) vectors. In this review, we will focus on the recent progress of viral vector mediated gene therapy in OUD, especially morphine tolerance and withdrawal.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Transtornos Relacionados ao Uso de Opioides/genética , Transtornos Relacionados ao Uso de Opioides/terapia , Animais , Dependovirus/genética , Humanos , Lentivirus/genética , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Simplexvirus/genética
8.
Pharmacogenomics ; 21(13): 903-917, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32757697

RESUMO

Aim: Heroin addiction is a chronic, relapsing disease that has genetic and environmental, including drug-induced, contributions. Stress influences the development of addictions. This study was conducted to determine if variants in stress-related genes are associated with opioid dependence (OD). Patients & methods: One hundred and twenty variants in 26 genes were analyzed in 597 Dutch subjects. Patients included 281 OD in methadone maintenance with or without heroin-assisted treatment and 316 controls. Results: Twelve SNPs in seven genes showed a nominally significant association with OD. Experiment-wise significant associations (p < 0.05) were found for three SNP pairs, through an interaction effect: NPY1R/GAL rs4691910/rs1893679, NPY1R/GAL rs4691910/rs3136541 and GALR1/GAL rs9807208/rs3136541. Conclusion: This study lends more evidence to previous reports of association of stress-related variants with heroin dependence.


Assuntos
Predisposição Genética para Doença/genética , Transtornos Relacionados ao Uso de Opioides/genética , Polimorfismo de Nucleotídeo Único/genética , Receptor Tipo 1 de Galanina/genética , Receptores de Neuropeptídeo Y/genética , Adulto , Estudos de Casos e Controles , Feminino , Genótipo , Haplótipos/genética , Heroína/uso terapêutico , Dependência de Heroína/genética , Humanos , Masculino , Metadona/uso terapêutico , Tratamento de Substituição de Opiáceos/métodos
9.
JAMA Psychiatry ; 77(10): 1072-1080, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32492095

RESUMO

Importance: With the current opioid crisis, it is important to improve understanding of the biological mechanisms of opioid use disorder (OUD). Objectives: To detect genetic risk variants for OUD and determine genetic correlations and causal association with OUD and other traits. Design, Setting, and Participants: A genome-wide association study of electronic health record-defined OUD in the Million Veteran Program sample was conducted, comprising 8529 affected European American individuals and 71 200 opioid-exposed European American controls (defined by electronic health record trajectory analysis) and 4032 affected African American individuals and 26 029 opioid-exposed African American controls. Participants were enrolled from January 10, 2011, to May 21, 2018, with electronic health record data for OUD diagnosis from October 1, 1999, to February 7, 2018. Million Veteran Program results and additional OUD case-control genome-wide association study results from the Yale-Penn and Study of Addiction: Genetics and Environment samples were meta-analyzed (total numbers: European American individuals, 10 544 OUD cases and 72 163 opioid-exposed controls; African American individuals, 5212 cases and 26 876 controls). Data on Yale-Penn participants were collected from February 14, 1999, to April 1, 2017, and data on Study of Addiction: Genetics and Environment participants were collected from 1990 to 2007. The key result was replicated in 2 independent cohorts: proxy-phenotype buprenorphine treatment in the UK Biobank and newly genotyped Yale-Penn participants. Genetic correlations between OUD and other traits were tested, and mendelian randomization analysis was conducted to identify potential causal associations. Main Outcomes and Measures: Main outcomes were International Classification of Diseases, Ninth Revision-diagnosed OUD or International Statistical Classification of Diseases and Related Health Problems, Tenth Revision-diagnosed OUD (Million Veteran Program), and DSM-IV-defined opioid dependence (Yale-Penn and Study of Addiction: Genetics and Environment). Results: A total of 114 759 individuals (101 016 men [88%]; mean [SD] age, 60.1 [12.8] years) were included. In 82 707 European American individuals, a functional coding variant (rs1799971, encoding Asn40Asp) in OPRM1 (µ-opioid receptor gene, the main biological target for opioid drugs; OMIM 600018) reached genome-wide significance (G allele: ß = -0.066 [SE = 0.012]; P = 1.51 × 10-8). The finding was replicated in 2 independent samples. Single-nucleotide polymorphism-based heritability of OUD was 11.3% (SE = 1.8%). Opioid use disorder was genetically correlated with 83 traits, including multiple substance use traits, psychiatric illnesses, cognitive performance, and others. Mendelian randomization analysis revealed the following associations with OUD: risk of tobacco smoking, depression, neuroticism, worry neuroticism subcluster, and cognitive performance. No genome-wide significant association was detected for African American individuals or in transpopulation meta-analysis. Conclusions and Relevance: This genome-wide meta-analysis identified a significant association of OUD with an OPRM1 variant, which was replicated in 2 independent samples. Post-genome-wide association study analysis revealed associated pleiotropic characteristics. Recruitment of additional individuals with OUD for future studies-especially those of non-European ancestry-is a crucial next step in identifying additional significant risk loci.


Assuntos
Transtornos Relacionados ao Uso de Opioides/genética , Receptores Opioides mu/análise , Idoso , Feminino , Estudo de Associação Genômica Ampla/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Receptores Opioides mu/sangue , Estados Unidos , United States Department of Veterans Affairs/organização & administração , United States Department of Veterans Affairs/estatística & dados numéricos
10.
Clin Epigenetics ; 12(1): 76, 2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32493461

RESUMO

BACKGROUND: A long-term opioid use has been associated with hypermethylation of the opioid receptor mu 1 (OPRM1) promoter. Very little is currently known about the early epigenetic response to therapeutic opioids. Here, we examine whether we can detect DNA methylation changes associated with a few days' use of prescribed opioids. Genome-wide DNA methylation was assayed in a cohort of 33 opioid-naïve participants who underwent standard dental surgery followed by opioid self-administration. Saliva samples were collected before surgery (visit 1), and at two postsurgery visits at 2.7 ± 1.5 days (visit 2), and 39 ± 10 days (visit 3) after the discontinuation of opioid analgesics. RESULTS: The perioperative methylome underwent significant changes over the three visits that were primarily due to postoperative inflammatory response and cell heterogeneity. To specifically examine the effect of opioids, we started with a candidate gene approach and evaluated 10 CpGs located in the OPRM1 promoter. There was a significant cross-sectional variability in opioid use, and for participants who self-administered the prescribed drugs, the total dosage ranged from 5-210 morphine milligram equivalent (MME). Participants were categorized by cumulative dosage into three groups: < 25 MME, 25-90 MME, and ≥ 90 MME. Using mixed-effects modeling, 4 CpGs had significant positive associations with opioid dose at two-tailed p value < 0.05, and overall, 9 of the 10 OPRM1 promoter CpGs showed the predicted higher methylation in the higher dose groups relative to the lowest dose group. After adjustment for age, cellular heterogeneity, and past tobacco use, the promoter mean methylation also had positive associations with cumulative MME (regression coefficient = 0.0002, one-tailed p value = 0.02) and duration of opioid use (regression coefficient = 0.003, one-tailed p value = 0.001), but this effect was significant only for visit 3. A preliminary epigenome-wide association study identified a significant CpG in the promoter of the RAS-related signaling gene, RASL10A, that may be predictive of opioid dosage. CONCLUSION: The present study provides evidence that the hypermethylation of the OPRM1 promoter is in response to opioid use and that epigenetic differences in OPRM1 and other sites are associated with a short-term use of therapeutic opioids.


Assuntos
Analgésicos Opioides/farmacologia , Metilação de DNA/efeitos dos fármacos , Transtornos Relacionados ao Uso de Opioides/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Adulto , Analgésicos Opioides/administração & dosagem , Estudos de Casos e Controles , Ilhas de CpG/genética , Epigênese Genética , Epigenoma/efeitos dos fármacos , Epigenoma/genética , Feminino , Estudo de Associação Genômica Ampla/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos Relacionados ao Uso de Opioides/metabolismo , Período Perioperatório , Variantes Farmacogenômicos/genética , Regiões Promotoras Genéticas/genética , Receptores Opioides mu/metabolismo , Saliva/metabolismo , Proteínas ras/efeitos dos fármacos , Proteínas ras/genética
11.
PLoS One ; 15(6): e0234549, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32555608

RESUMO

Methadone is a synthetic opioid used as maintenance treatment for patients addicted to heroin. Skin irritation is one of the adverse events caused by opioid use. 344 methadone maintenance treatment (MMT) patients were recruited with records and measurements on methadone dose, plasma methadone concentrations, and treatment emergent symptom scales (TESS). 15 patients reported with skin irritation. Five SNPs located within the NECTIN4 genetic region were genotyped. The NECTIN4 gene within the adherens junction interaction pathway was associated with methadone dose in pathway-based genome wide association analyses (P = 0.0008). Three highly-linked SNPs, rs11265549, rs3820097, and rs4656978, were significantly associated with methadone dose (P = 0.0003), plasma concentrations of R,S-methadone (P = 0.0004) and TNF-α (P = 0.010) in all 344 MMT patients, and with self-report skin irritation symptom scores (P = 0.010) in the 15 MMT patients who reported with skin irritation. To identify the possible roles of plasma level of Nectin-4 in the responses to MMT and opioid use, additional age- and gender-matched 51 controls and 83 methadone-free abstinent former heroin users were recruited. Plasma level of Nectin-4 was the highest in MMT patients among the three groups. The results suggest involvement of genetic variants on NECTIN4 in methadone dose. Plasma Nectin-4 level is likely an indicator for continued use of opioids.


Assuntos
Moléculas de Adesão Celular/genética , Dependência de Heroína/genética , Metadona/administração & dosagem , Transtornos Relacionados ao Uso de Opioides/genética , Adulto , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/efeitos adversos , Moléculas de Adesão Celular/sangue , Feminino , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Dependência de Heroína/sangue , Dependência de Heroína/tratamento farmacológico , Dependência de Heroína/patologia , Humanos , Masculino , Metadona/efeitos adversos , Metadona/sangue , Tratamento de Substituição de Opiáceos/métodos , Transtornos Relacionados ao Uso de Opioides/sangue , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/patologia
14.
Neurosci Lett ; 704: 100-105, 2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-30936032

RESUMO

The dynorphin/kappa opioid receptor (Dyn/KOR) system is involved in reward processing and dysphoria/anhedonia. Exposure to mu-opioid receptor agonists such as heroin increases expression of the prodynorphin gene (PDYN) in the brain. In this study in a Caucasian cohort, we examined the association of the functional PDYN 68-bp repeat polymorphism with opioid use disorders. In this case-control study, 554 subjects with Caucasian ancestry (142 healthy controls, 153 opioid-exposed, but never opioid dependent, NOD, and 259 with an opioid dependence diagnosis, OD) were examined for association of the PDYN 68-bp repeats with the diagnosis of opioid dependence (DSM-IV criteria), with a dimensional measure of heroin exposure (KMSK scale), and age trajectory parameters of heroin use (age of heroin first use, and age of onset of heaviest use). The PDYN 68-bp repeat genotype (classified as: "short-short" [SS], "long-long" [LL], and "short-long" [SL], based on the number of repeats) was not associated with categorical opioid dependence diagnoses. However, the LL genotype was associated with later age of first heroin use than the SS + SL genotype (19 versus 18 years; p < 0.01). This was also confirmed by a significant positive correlation between the number of repeats and the age of first use of heroin, in volunteers with OD (Spearman r = 0.16; p = 0.01). This suggests that the functional PDYN 68-bp repeat genotype is associated with the age of first use of heroin in Caucasians diagnosed with opioid dependence.


Assuntos
Encefalinas/genética , Transtornos Relacionados ao Uso de Opioides/genética , Precursores de Proteínas/genética , Adulto , Estudos de Casos e Controles , Feminino , Estudos de Associação Genética , Genótipo , Dependência de Heroína/diagnóstico , Dependência de Heroína/etnologia , Dependência de Heroína/genética , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos Relacionados ao Uso de Opioides/diagnóstico , Transtornos Relacionados ao Uso de Opioides/etnologia , Polimorfismo Genético , Regiões Promotoras Genéticas , População Branca
15.
Pharmacogenomics ; 20(5): 331-341, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30983500

RESUMO

Aim: To determine if selected serotonergic and noradrenergic gene variants are associated with heroin addiction. Subjects & methods: A total of 126 variants in 19 genes in subjects with Dutch European ancestry from The Netherlands. Subjects included 281 opioid-dependent volunteers in methadone maintenance or heroin-assisted treatment, 163 opioid-exposed but not opioid-dependent volunteers who have been using illicit opioids but never became opioid-dependent and 153 healthy controls. Results: Nominal associations were indicated for 20 variants in six genes including an experiment-wise significant association from the combined effect of three SLC18A2 SNPs (rs363332, rs363334 and rs363338) with heroin dependence (pfinal = 0.047). Conclusion: Further studies are warranted to confirm and elucidate the role of these variants in the vulnerability to opioid addiction.


Assuntos
Transtornos Relacionados ao Uso de Opioides/genética , Polimorfismo de Nucleotídeo Único , Proteínas Vesiculares de Transporte de Monoamina/genética , Adulto , Analgésicos Opioides/administração & dosagem , Estudos de Casos e Controles , Feminino , Predisposição Genética para Doença , Dependência de Heroína/tratamento farmacológico , Dependência de Heroína/genética , Humanos , Masculino , Metadona/uso terapêutico , Pessoa de Meia-Idade , Países Baixos , Tratamento de Substituição de Opiáceos , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Testes Farmacogenômicos , Fatores de Risco , População Branca/genética
16.
PLoS Genet ; 15(4): e1008060, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31022172

RESUMO

The promise of personalized genomic medicine is that knowledge of a person's gene sequences and activity will facilitate more appropriate medical interventions, particularly drug prescriptions, to reduce the burden of disease. Early successes in oncology and pediatrics have affirmed the power of positive diagnosis and are mostly based on detection of one or a few mutations that drive the specific pathology. However, genetically more complex diseases require the development of polygenic risk scores (PRSs) that have variable accuracy. The rarity of events often means that they have necessarily low precision: many called positives are actually not at risk, and only a fraction of cases are prevented by targeted therapy. In some situations, negative prediction may better define the population at low risk. Here, I review five conditions across a broad spectrum of chronic disease (opioid pain medication, hypertension, type 2 diabetes, major depression, and osteoporotic bone fracture), considering in each case how genetic prediction might be used to target drug prescription. This leads to a call for more research designed to evaluate genetic likelihood of response to therapy and a call for evaluation of PRS, not just in terms of sensitivity and specificity but also with respect to potential clinical efficacy.


Assuntos
Herança Multifatorial , Medicina de Precisão/métodos , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/genética , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/genética , Feminino , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/genética , Masculino , Mutação , Transtornos Relacionados ao Uso de Opioides/genética , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Osteoporose/genética , Osteoporose/prevenção & controle , Testes Farmacogenômicos/métodos , Medicina Preventiva/métodos , Fatores de Risco
17.
Sci Rep ; 9(1): 4660, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30874594

RESUMO

There is currently an epidemic of opioid use, overdose, and dependence in the United States. Although opioid dependence (OD) is more prevalent in men, opioid relapse and fatal opioid overdoses have recently increased at a higher rate among women. Epigenetic mechanisms have been implicated in the etiology of OD, though most studies to date have used candidate gene approaches. We conducted the first epigenome-wide association study (EWAS) of OD in a sample of 220 European-American (EA) women (140 OD cases, 80 opioid-exposed controls). DNA was derived from whole blood samples and EWAS was implemented using the Illumina Infinium HumanMethylationEPIC array. To identify differentially methylated CpG sites, we performed an association analysis adjusting for age, estimates of cell proportions, smoking status, and the first three principal components to correct for population stratification. After correction for multiple testing, association analysis identified three genome-wide significant differentially methylated CpG sites mapping to the PARG, RERE, and CFAP77 genes. These genes are involved in chromatin remodeling, DNA binding, cell survival, and cell projection. Previous genome-wide association studies have identified RERE risk variants in association with psychiatric disorders and educational attainment. DNA methylation age in the peripheral blood did not differ between OD subjects and opioid-exposed controls. Our findings implicate epigenetic mechanisms in OD and, if replicated, identify possible novel peripheral biomarkers of OD that could inform the prevention and treatment of the disorder.


Assuntos
Epigênese Genética/genética , Transtornos Relacionados ao Uso de Opioides/genética , Adulto , Biomarcadores/sangue , Ilhas de CpG/genética , DNA/sangue , Metilação de DNA/genética , Epigenômica/métodos , Feminino , Estudo de Associação Genômica Ampla/métodos , Humanos , Pessoa de Meia-Idade , Estados Unidos , População Branca/genética
18.
Sci Rep ; 9(1): 1534, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30733491

RESUMO

Opioid abuse is now the most common cause of accidental death in the US. Although opioids and most other drugs of abuse acutely increase signaling mediated by midbrain dopamine (DA)-synthesizing neurons, little is known about long-lasting changes in DA cells that may contribute to continued opioid abuse, craving, and relapse. A better understanding of the molecular and cellular bases of opioid abuse could lead to advancements in therapeutics. This study comprises, to our knowledge, the first unbiased examination of genome-wide changes in midbrain gene expression associated with human opioid abuse. Our analyses identified differentially expressed genes and distinct gene networks associated with opioid abuse, specific genes with predictive capability for subject assignment to the opioid abuse cohort, and genes most similarly affected in chronic opioid and cocaine abusers. We also identified differentially expressed long noncoding RNAs capable of regulating known drug-responsive protein-coding genes. Opioid-regulated genes identified in this study warrant further investigation as potential biomarkers and/or therapeutic targets for human substance abuse.


Assuntos
Biomarcadores/metabolismo , Cocaína/farmacologia , Redes Reguladoras de Genes , Mesencéfalo/metabolismo , Transtornos Relacionados ao Uso de Opioides/patologia , RNA Longo não Codificante/metabolismo , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Área Sob a Curva , Estudos de Casos e Controles , Humanos , Concentração de Íons de Hidrogênio , Mesencéfalo/química , Mesencéfalo/efeitos dos fármacos , Pessoa de Meia-Idade , Inibidor de NF-kappaB alfa/genética , Inibidor de NF-kappaB alfa/metabolismo , Transtornos Relacionados ao Uso de Opioides/genética , Transtornos Relacionados ao Uso de Opioides/metabolismo , Curva ROC
19.
J Cell Physiol ; 234(3): 3058-3066, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30146703

RESUMO

The chronic use of opioids leads to tolerance, psychological, and physical dependence that limits their use as an effective long-term pain control. Several studies have shown that mammalian target of rapamycin (mTOR) plays a crucial role in the development of opioid tolerance. Metformin activates 5' adenosine monophosphate-activated protein kinase (AMPK) which directly suppresses the mTOR complex 1 signaling pathway. On the other hand, metformin can also inhibit mTOR directly and in an AMPK-independent manner. Thus, in the current study, we aimed to investigate the effects of metformin on the development of morphine and/or methadone-induced tolerance in human glioblastoma (T98G) cell line. We examined the effects of chronic treatment of morphine and/or methadone in the presence or absence of metformin with or without AMPK inhibitor (dorsomorphin hydrochloride) on levels of nitric oxide and cyclic adenosine monophosphate (cAMP), phosphorylated and dephosphorylated ribosomal protein S6 kinase ß-1 (S6K1) and 4E-binding protein 1 (4E-BP1) in T98G cells. Pretreatment of cells with metformin (40 µM) with or without AMPK inhibitor (dorsomorphin hydrochloride; 1 µM) before adding of morphine (2.5 µM) or methadone (1 µM) revealed a protective effects on the development of opioid tolerance. Prior administration of metformin reversed the elevation of nitric oxide levels induced by morphine (p < 0.001) and methadone (p < 0.001) and also prevented the raise of cAMP levels induced by morphine in T98G cells (p < 0.05). Contribution of mTOR signaling pathway in metformin-induced effect was shown by the inhibition of phosphorylation of S6K1 and 4E-BP1, the downstream targets of mTOR. mTOR activation suppresses opioid-induced antinociception, and its activity has also been increased during opioid tolerance.


Assuntos
Metformina/farmacologia , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Proteínas Quinases/genética , Serina-Treonina Quinases TOR/genética , Quinases Proteína-Quinases Ativadas por AMP , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacologia , Animais , Linhagem Celular Tumoral , AMP Cíclico/genética , Tolerância a Medicamentos/genética , Fatores de Iniciação em Eucariotos/genética , Glioblastoma/tratamento farmacológico , Humanos , Metadona/efeitos adversos , Metadona/farmacologia , Morfina/efeitos adversos , Morfina/farmacologia , Transtornos Relacionados ao Uso de Opioides/genética , Transtornos Relacionados ao Uso de Opioides/patologia , Fosforilação/efeitos dos fármacos , Proteínas Quinases/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Transdução de Sinais/efeitos dos fármacos
20.
Basic Clin Pharmacol Toxicol ; 124(4): 439-448, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30549211

RESUMO

The threats involved in the long-term opioid treatment of chronic non-cancer pain (CNCP) have increased notably. Strategies to identify at-risk patients are important because there is no clear evidence showing which screening or deprescription programmes are appropriate. Our aim was to evaluate the evidence provided by pharmacogenetics applied to predict an analgesic toxicity profile in prescription opioid use disorder (POUD) patients participating in an opioid deprescription programme. Pharmacogenetic markers were analysed in an observational, prospective deprescription programme for POUD patients (n = 88) treated for CNCP. It consisted of monitoring visits (baseline, follow-up and final), opioid rotation or discontinuation and the recording of adverse events and suspected adverse drug reactions (ADRs). Variants in OPRM1 (A118G), ABCB1 (C3435T), COMT (G472A), OPRD1 (T921C) and ARRB2 (C8622T) genes were tested by real-time PCR. Ethics committee approved the study. Wild-type OPRM1-AA genotype carriers reported a significantly higher number of adverse events than OPRM1-AG/GG (median [p25-75], 7 [5-11] vs 5 [3-9]), particularly gastrointestinal system events (90% vs 63%) such as nausea (33% vs 0%). Suspected ADRs (affecting 17% of the patients) were three times higher in males than in females (30% vs 11%). The deprescription programme was effective and safe, and it achieved a significant progressive reduction in the morphine equivalent daily dose, strong opioids and other analgesics' use, without causing any changes in pain intensity or opiate abstinence syndrome. OPRM1 gene polymorphisms could identify the risk of gastrointestinal adverse events in POUD patients. Deprescription programmes including pharmacogenetic analysis should be considered during the follow-up of this population.


Assuntos
Analgésicos Opioides/administração & dosagem , Desprescrições , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Farmacogenética , Analgésicos Opioides/efeitos adversos , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos Relacionados ao Uso de Opioides/genética , Estudos Prospectivos , Receptores Opioides mu/genética , Fatores Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA