Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Sci Rep ; 13(1): 5827, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-37037889

RESUMO

Although opioids are necessary for the treatment of acute pain, cancer pain, and palliative care, opioid abuse is a serious threat to society. Heroin (Diacetylmorphine) is the most commonly abused opioid, and it can have a variety of effects on the body's tissues and organs, including the well-known gastrointestinal depression and respiratory depression; however, there is little known about the effects of diacetylmorphine on cardiac damage. Here, we demonstrate that diacetylmorphine induces abnormal electrocardiographic changes in rats and causes damage to cardiomyocytes in vitro by an underlying mechanism of increased autophosphorylation of CaMKII and concomitant regulation of myocardial contractile protein TPM1 and MYOM2 protein expression. The CaMKII inhibitor KN-93 was first tested to rescue the toxic effects of heroin on cardiomyocytes in vitro and the abnormal ECG changes caused by heroin in SD rats, followed by the TMT relative quantitative protein technique to analyze the proteome changes. Diacetylmorphine causes increased phosphorylation at the CaMKII Thr287 site in myocardium, resulting in increased autophosphorylation of CaMKII and subsequent alterations in myocardial contractile proteins, leading to myocardial rhythm abnormalities. These findings provide a theoretical basis for the treatment and prevention of patients with arrhythmias caused by diacetylmorphine inhalation and injection.


Assuntos
Arritmias Cardíacas , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Heroína , Transtornos Relacionados ao Uso de Opioides , Animais , Ratos , Analgésicos Opioides , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Heroína/toxicidade , Miócitos Cardíacos/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Fosforilação , Ratos Sprague-Dawley , Tropomiosina/metabolismo
2.
J Adv Res ; 54: 43-57, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-36716956

RESUMO

BACKGROUND: Murine Nischarin and its human homolog IRAS are scaffold proteins highly expressed in the central nervous system (CNS). Nischarin was initially discovered as a tumor suppressor protein, and recent studies have also explored its potential value in the CNS. Research on IRAS has largely focused on its effect on opioid dependence. Although the role of Nischarin/IRAS in the physiological function and pathological process of the CNS has gradually attracted attention and the related research results are expected to be applied in clinical practice, there is no systematic review of the role and mechanisms of Nischarin/IRAS in the CNS so far. AIM OF REVIEW: This review will systematically analyze the role and mechanism of Nischarin/IRAS in the CNS, and provide necessary references and possible targets for the treatment of neurological diseases, thereby broadening the direction of Nischarin/IRAS research and facilitating clinical translation. KEY SCIENTIFIC CONCEPTS OF REVIEW: The pathophysiological processes affected by dysregulation of Nischarin/IRAS expression in the CNS are mainly introduced, including spinal cord injury (SCI), opioid dependence, anxiety, depression, and autism. The molecular mechanisms such as factors regulating Nischarin/IRAS expression and signal transduction pathways regulated by Nischarin/IRAS are systematically summarized. Finally, the clinical application of Nischarin/IRAS has been prospected.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Transtornos Relacionados ao Uso de Opioides , Camundongos , Humanos , Animais , Receptores de Imidazolinas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Transporte/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Transdução de Sinais
3.
Neuropharmacology ; 205: 108925, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34921830

RESUMO

The abuse of oral formulations of prescription opioids has precipitated the current opioid epidemic. We developed an oral oxycodone consumption model consisting of a limited access (4 h) two-bottle choice drinking in the dark (TBC-DID) paradigm and quantified dependence with naloxone challenge using mice of both sexes. We also assessed neurobiological correlates of withdrawal and dependence elicited via oral oxycodone consumption using immunohistochemistry for DeltaFosB (ΔFosB), a transcription factor described as a molecular marker for drug addiction. Neither sex developed a preference for the oxycodone bottle, irrespective of oxycodone concentration, bottle position or prior water restriction. Mice that volitionally consumed oxycodone exhibited hyperlocomotion in an open field test and supraspinal but not spinally-mediated antinociception. Both sexes also developed robust, dose-dependent levels of opioid withdrawal that was precipitated by the opioid antagonist naloxone. Oral oxycodone consumption followed by naloxone challenge led to mesocorticolimbic region-dependent increases in the number of ΔFosB expressing cells. Naloxone-precipitated withdrawal jumps, but not the oxycodone bottle % preference, was positively correlated with the number of ΔFosB expressing cells specifically in the nucleus accumbens shell. Thus, limited access oral consumption of oxycodone produced physical dependence and increased ΔFosB expression despite the absence of opioid preference. Our TBC-DID paradigm allows for the study of oral opioid consumption in a simple, high-throughput manner and elucidates the underlying neurobiological substrates that accompany opioid-induced physical dependence.


Assuntos
Analgésicos Opioides/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Transtornos Relacionados ao Uso de Opioides , Oxicodona/farmacologia , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Síndrome de Abstinência a Substâncias , Analgésicos Opioides/administração & dosagem , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Locomoção/efeitos dos fármacos , Camundongos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Nociceptividade/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Oxicodona/administração & dosagem , Proteínas Proto-Oncogênicas c-fos/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/fisiopatologia
4.
Pharmacol Biochem Behav ; 212: 173294, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34752798

RESUMO

Over the past two decades, opioid abuse has risen especially among women. In both sexes hippocampal neural circuits involved in associative memory formation and encoding of motivational incentives are critically important in the transition from initial drug use to drug abuse/dependence. Opioid circuits, particularly the mossy fiber pathway, are crucial for associative memory processes important for addiction. Our anatomical studies, especially those utilizing electron microscopic immunocytochemistry, have provided unique insight into sex differences in the distribution of opioid peptides and receptors in specific hippocampal circuits and how these distributions are altered following stress and oxycodone-associative learning processes. Here we review the hippocampal opioid system in rodents with respect to ovarian hormones effects and baseline sex differences then sex differences following acute and chronic stress. Next, we review sex differences in the hippocampal opioid system in unstressed and chronically stressed rats following oxycodone conditioned place preference. We show that opioid peptides and receptors are distributed within hippocampal circuits in females with elevated estrogen states in a manner that would enhance sensitivity to endogenous and exogenous opioids. Moreover, chronic stress primes the opioid system in females in a manner that would promote opioid-associative learning processes. In contrast, chronic stress has limited effects on the opioid system in males and reduces its capacity to support opioid-mediated learning processes. Interestingly, acute stress appears to prime males for opioid associative learning. On a broader scale the findings highlighted in this review have important implications in understanding sex differences in opioid drug use and abuse.


Assuntos
Hipocampo/efeitos dos fármacos , Aprendizagem/efeitos dos fármacos , Transtornos Relacionados ao Uso de Opioides/metabolismo , Oxicodona/farmacologia , Receptores Opioides/metabolismo , Estresse Psicológico/metabolismo , Analgésicos Opioides/farmacologia , Animais , Condicionamento Clássico , Feminino , Hipocampo/metabolismo , Masculino , Microscopia Eletrônica/métodos , Neurônios/metabolismo , Peptídeos Opioides/farmacologia , Ratos , Receptores de Estrogênio/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Caracteres Sexuais
5.
J Biochem Mol Toxicol ; 34(10): e22559, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33034947

RESUMO

To date, not much study has been done to investigate the mitochondrial DNA (mtDNA) copy number as the potential biomarker for opium exposure. Here, we conducted a cross-sectional study to determine the relative mtDNA content as the potential biomarker for opium exposure. Quantitative real-time PCR was performed to investigate the mtDNA copy number variation across 205 individuals, including blood samples of 45 opium users, 41 cigarette users, 47 dual users, and 72 never users of any product. We found a significantly higher mtDNA content among the opium-only users (adjusted OR: 3.21; 95% CI: [1.34, 7.66]; P = .009) and dual users (adjusted OR: 2.64; 95% CI: [1.15, 6.1]; P = .02) compared to that in never users even after adjustment for confounding factors, age, and sex. Discordantly, analysis of mitochondrial DNA in cigarette smokers revealed an indirect association between cigarette smoking and mtDNA content although it was not statistically significant. The reason behind the increased mitochondrial DNA is unclear. The possible hypothesis is that there might be a way to compensate for the oxidative damage induced by opium consumption. Taken together, our findings indicated that the mtDNA copy number may alter during opium exposure. Since changes in the mitochondrial DNA copy number was associated with the etiology of many diseases including cancer, further investigations on the mtDNA copy number may shed light on the carcinogenicity of opium consumption and means for early detection among the populations who have been exposed to opium and its products.


Assuntos
DNA Mitocondrial/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Adolescente , Adulto , Idoso , Biomarcadores/metabolismo , Estudos Transversais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Adulto Jovem
6.
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
7.
Nicotine Tob Res ; 22(10): 1923-1927, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-32303761

RESUMO

INTRODUCTION: Pregnant women exposed chronically to opioids smoked more cigarettes per day (CPD) and had a higher nicotine metabolite ratio (NMR), 3-hydroxycotinine/cotinine, a biomarker of nicotine metabolism and clearance, than those not receiving opioids. We examined CPD and NMR in a group of pregnant smokers, a quarter of whom were receiving opioid agonist therapy (OAT). AIMS AND METHODS: Pregnant smokers recruited to participate in a placebo-controlled trial of bupropion for smoking cessation provided a blood sample for measurement of NMR. RESULTS: Half (52.4%) of the 124 women with NMR data were African American. OAT-treated women (n = 34, 27.4%; 27 receiving methadone and 7 buprenorphine) were more likely to be white (79% vs. 30%, p < .001) and to have a lower mean PHQ-9 total score (2.91 [SD = 2.83] vs. 4.83 [SD = 3.82], p = .007). OAT-treated women reported smoking more CPD (9.50 [SD = 5.26] vs. 7.20 [SD = 3.65], p = .005) and had higher NMR (0.78 [SD = 0.36] vs. 0.56 [SD = 0.25], p = .001) than the non-OAT-treated group. In a linear regression analysis adjusting for race, depression severity, and CPD, NMR was greater in the OAT group (p = .025), among whom the daily methadone-equivalent dosage correlated with NMR (Spearman's ρ = 0.49, p = .003). CONCLUSIONS: Consistent with the findings of Oncken et al. (2019), we found that OAT smokers smoked more and had higher NMR than non-OAT smokers. As higher NMR is associated with a reduced likelihood of smoking cessation, the effects on NMR of both pregnancy and OAT could contribute to a lower smoking cessation rate in pregnant smokers receiving chronic opioid therapy. IMPLICATIONS: We replicated the finding that the NMR is significantly greater among pregnant smokers receiving OAT than those not receiving this treatment for opioid use disorder. Furthermore, we found that the dosage of the OAT was significantly associated with the NMR level. These findings may contribute to a poorer response to smoking cessation treatment in pregnant women treated with OAT, particularly those receiving high-dose therapy, and raise the question of whether novel approaches are needed to treat smoking in this subgroup of pregnant smokers.


Assuntos
Cotinina , Tratamento de Substituição de Opiáceos , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Complicações na Gravidez/tratamento farmacológico , Fumar/tratamento farmacológico , Analgésicos Opioides/agonistas , Bupropiona/uso terapêutico , Cotinina/análogos & derivados , Cotinina/sangue , Cotinina/metabolismo , Feminino , Humanos , Metadona/uso terapêutico , Nicotina/sangue , Nicotina/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Gravidez , Complicações na Gravidez/metabolismo , Abandono do Hábito de Fumar
8.
Hum Exp Toxicol ; 39(6): 808-815, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32009467

RESUMO

CONTEXT: Heavy metals, including thallium and lead, are introduced to illicit drug users' body as a result of using drugs such as cocaine and heroin. OBJECTIVE: This study aimed to determine urine, blood, and hair thallium (Tl) concentrations in illicit opioid users along with the relevant clinical signs and symptoms consistent with thallotoxicosis and to compare them with the corresponding variables in the control non-opioid user group. MATERIALS AND METHODS: This case-control study was conducted on 50 illicit opioid users who had abused opioids continuously for more than a year, referred to Amirie Drug Abuse Treatment Clinic in Kashan, Iran. The control group included 50 non-opioid users. Thallium concentrations in urine, blood, and hair were assessed in both groups (n = 100) using electrothermal (graphite furnace) atomic absorption spectrometry (ET AAS, GF AAS). RESULTS: In the studied group, the median (interquartile range) concentrations of thallium in urine, blood, and hair were 54.8 ± 79.9 µg/L, 14.5 ± 11.1 µg/L, and 5.4 ± 3.7 µg/g, respectively; these values were 4.8 ± 5.2 µg/L, 2.5 ± 2.4 µg/L, and 1.4 ± 1.1 µg/g, respectively, in the control group. There were significant differences in urine, blood, and hair thallium concentrations between the study group and the control group (p < 0.001). There were significant correlations between duration of illicit opioid use and urine thallium concentrations (r = 0.394, p = 0.005) and hair thallium concentrations (r = 0.293, p = 0.039), but not with blood thallium concentrations (r = 0.246, p = 0.085). Urine and blood thallium concentrations of illicit opioid users with clinical signs and symptoms consistent with thallotoxicosis of weakness (p = 0.01), depression (p = 0.03), and headache (p = 0.03) were higher than users without these problems. DISCUSSION AND CONCLUSION: The results of the study showed that thallium concentrations in urine, blood, and hair in illicit opioid users were significantly higher than the comparable concentrations in the control group. This can be due to the use of illicit opioids adulterated with thallium. Also, this study showed long-term illicit opioid use may lead to thallium exposure. In addition, cigarette smoking was associated with increased thallium exposure.


Assuntos
Cabelo/química , Transtornos Relacionados ao Uso de Opioides , Tálio , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Analgésicos Opioides/toxicidade , Estudos de Casos e Controles , Feminino , Heroína/toxicidade , Humanos , Drogas Ilícitas/toxicidade , Irã (Geográfico)/epidemiologia , Masculino , Pessoa de Meia-Idade , Transtornos Relacionados ao Uso de Opioides/sangue , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Transtornos Relacionados ao Uso de Opioides/metabolismo , Transtornos Relacionados ao Uso de Opioides/urina , Ópio/toxicidade , Tálio/análise , Tálio/sangue , Tálio/toxicidade , Tálio/urina , Adulto Jovem
9.
AIDS ; 34(2): 177-188, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31687981

RESUMO

BACKGROUND: Opioid-use disorders (OUD) and hepatitis C or B co-infection (HEP) are common among people living with HIV (PLHIV). The impact of OUD on innate and adaptive immunity among PLHIV with and without HEP is unknown. OBJECTIVES: To investigate the impact of OUD on monocyte and T-cell phenotypes, cytokine responses to lipopolysaccharide (LPS) and phytohemagglutinin (PHA), and plasma inflammatory markers, among PLHIV with and without HEP. METHODS: Cross-sectional study enrolling PLHIV receiving ART, with and without OUD. Flow cytometry determined monocyte and T-cell phenotypes; LPS and PHA-induced cytokine production was assessed following LPS and PHA stimulation by multiplex cytokine array; plasma IL-6, soluble CD163, and soluble CD14 were measured by ELISA. RESULTS: Twenty-two PLHIV with OUD and 37 PLHIV without OUD were included. PLHIV with OUD exhibited higher frequencies of intermediate (CD14CD16) and nonclassical (CD14CD16) monocytes when compared with PLHIV without OUD (P = 0.0025; P = 0.0001, respectively), regardless of HEP co-infection. Soluble CD163 and monocyte cell surface CD163 expression was increased among PLHIV with OUD and HEP, specifically. Regardless of HEP co-infection, PLHIV with OUD exhibited reduced production of IL-10, IL-8, IL-6, IL-1alpha, and TNF-alpha in response to LPS when compared with PLHIV without OUD; PHA-induced production of IL-10, IL-1alpha, IL-1beta, IL-6, and TNF-alpha were also reduced among individuals with OUD. CONCLUSION: OUD among PLHIV are associated with altered monocyte phenotypes and a dysregulated innate cytokine response. Defining underlying mechanisms of opioid-associated innate immune dysregulation among PLHIV should be prioritized to identify optimal OUD treatment strategies.


Assuntos
Antígenos CD/sangue , Antígenos de Diferenciação Mielomonocítica/sangue , Citocinas/metabolismo , Infecções por HIV/metabolismo , Monócitos/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Receptores de Superfície Celular/sangue , Adulto , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Biomarcadores/metabolismo , Estudos Transversais , Feminino , Citometria de Fluxo , Infecções por HIV/sangue , Humanos , Interleucina-10/metabolismo , Interleucina-1beta , Masculino , Pessoa de Meia-Idade , Transtornos Relacionados ao Uso de Opioides/sangue , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptores de Superfície Celular/metabolismo , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/metabolismo
10.
Cell Rep ; 29(4): 946-960.e2, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31644915

RESUMO

Opioids are powerful addictive agents that alter dopaminergic influence on reward signaling in medium spiny neurons (MSNs) of the nucleus accumbens. Repeated opioid exposure triggers adaptive changes, shifting reward valuation to the allostatic state underlying tolerance. However, the cellular substrates and molecular logic underlying such allostatic changes are not well understood. Here, we report that the plasticity of dopamine-induced cyclic AMP (cAMP) signaling in MSNs serves as a cellular substrate for drug-induced allostatic adjustments. By recording cAMP responses to optically evoked dopamine in brain slices from mice subjected to various opioid exposure paradigms, we define profound neuronal-type-specific adaptations. We find that opioid exposure pivots the initial hyper-responsiveness of D1-MSNs toward D2-MSN dominance as dependence escalates. Presynaptic dopamine transporters and postsynaptic phosphodiesterases critically enable cell-specific adjustments of cAMP that control the balance between opponent D1-MSN and D2-MSN channels. We propose a quantitative model of opioid-induced allostatic adjustments in cAMP signal strength that balances circuit activity.


Assuntos
Adaptação Fisiológica , Corpo Estriado/metabolismo , AMP Cíclico/metabolismo , Dopamina/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Transmissão Sináptica , Analgésicos Opioides/farmacologia , Animais , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/fisiopatologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/fisiologia , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Diester Fosfórico Hidrolases/metabolismo
11.
Neuropharmacology ; 158: 107609, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31009632

RESUMO

Substance use disorders (SUD) are serious public health problems worldwide. Although significant progress has been made in understanding the neurobiology of drug reward and the transition to addiction, effective pharmacotherapies for SUD remain limited and a majority of drug users relapse even after a period of treatment. The United States Food and Drug Administration (FDA) has approved several medications for opioid, nicotine, and alcohol use disorders, whereas none are approved for the treatment of cocaine or other psychostimulant use disorders. The medications approved by the FDA for the treatment of SUD can be divided into two major classes - agonist replacement therapies, such as methadone and buprenorphine for opioid use disorders (OUD), nicotine replacement therapy (NRT) and varenicline for nicotine use disorders (NUD), and antagonist therapies, such as naloxone for opioid overdose and naltrexone for promoting abstinence. In the present review, we primarily focus on the pharmacological rationale of agonist replacement strategies in treatment of opioid dependence, and the potential translation of this rationale to new therapies for cocaine use disorders. We begin by describing the neural mechanisms underlying opioid reward, followed by preclinical and clinical findings supporting the utility of agonist therapies in the treatment of OUD. We then discuss recent progress of agonist therapies for cocaine use disorders based on lessons learned from methadone and buprenorphine. We contend that future studies should identify agonist pharmacotherapies that can facilitate abstinence in patients who are motivated to quit their illicit drug use. Focusing on those that are able to achieve abstinence from cocaine will provide a platform to broaden the effectiveness of medication and psychosocial treatment strategies for this underserved population. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.


Assuntos
Analgésicos Opioides/uso terapêutico , Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Inibidores da Captação de Dopamina/uso terapêutico , Tratamento de Substituição de Opiáceos , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Buprenorfina/uso terapêutico , Estimulantes do Sistema Nervoso Central/uso terapêutico , Dextroanfetamina/uso terapêutico , Proteínas da Membrana Plasmática de Transporte de Dopamina/antagonistas & inibidores , Desenvolvimento de Medicamentos , Humanos , Metadona/uso terapêutico , Metilfenidato/uso terapêutico , Modafinila/uso terapêutico , Agonistas Nicotínicos/uso terapêutico , Transtornos Relacionados ao Uso de Opioides/metabolismo , Oxalatos , Piperazinas , Receptores Opioides mu/agonistas , Tabagismo/tratamento farmacológico , Tropanos/uso terapêutico , Vareniclina/uso terapêutico
12.
Peptides ; 115: 51-58, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30890355

RESUMO

Each year, over 50 million Americans suffer from persistent pain, including debilitating headaches, joint pain, and severe back pain. Although morphine is amongst the most effective analgesics available for the management of severe pain, prolonged morphine treatment results in decreased analgesic efficacy (i.e., tolerance). Despite significant headway in the field, the mechanisms underlying the development of morphine tolerance are not well understood. The midbrain ventrolateral periaqueductal gray (vlPAG) is a primary neural substrate for the analgesic effects of morphine, as well as for the development of morphine tolerance. A growing body of literature indicates that activated glia (i.e., microglia and astrocytes) facilitate pain transmission and oppose morphine analgesia, making these cells important potential targets in the treatment of chronic pain. Morphine affects glia by binding to the innate immune receptor toll-like receptor 4 (TLR4), leading to the release of proinflammatory cytokines and opposition of morphine analgesia. Despite the established role of the vlPAG as an integral locus for the development of morphine tolerance, most studies have examined the role of glia activation within the spinal cord. Additionally, the role of TLR4 in the development of tolerance has not been elucidated. This review attempts to summarize what is known regarding the role of vlPAG glia and TLR4 in the development of morphine tolerance. These data, together, provide information about the mechanism by which central nervous system glia regulate morphine tolerance, and identify a potential therapeutic target for the enhancement of analgesic efficacy in the clinical treatment of chronic pain.


Assuntos
Analgésicos Opioides/efeitos adversos , Dor Crônica/tratamento farmacológico , Tolerância a Medicamentos , Mediadores da Inflamação/metabolismo , Morfina/efeitos adversos , Transtornos Relacionados ao Uso de Opioides/metabolismo , Analgésicos Opioides/uso terapêutico , Dor Crônica/metabolismo , Humanos , Morfina/uso terapêutico , Substância Cinzenta Periaquedutal/metabolismo , Medula Espinal/metabolismo , Receptor 4 Toll-Like/metabolismo
13.
Neurosci Bull ; 35(3): 486-496, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30721395

RESUMO

MicroRNA-132 (miR-132), a small RNA that regulates gene expression, is known to promote neurogenesis in the embryonic nervous system and adult brain. Although exposure to psychoactive substances can increase miR-132 expression in cultured neural stem cells (NSCs) and the adult brain of rodents, little is known about its role in opioid addiction. So, we set out to determine the effect of miR-132 on differentiation of the NSCs and whether this effect is involved in opioid addiction using the rat morphine self-administration (MSA) model. We found that miR-132 overexpression enhanced the differentiation of NSCs in vivo and in vitro. Similarly, specific overexpression of miR-132 in NSCs of the adult hippocampal dentate gyrus (DG) during the acquisition stage of MSA potentiated morphine-seeking behavior. These findings indicate that miR-132 is involved in opioid addiction, probably by promoting the differentiation of NSCs in the adult DG.


Assuntos
Diferenciação Celular , Giro Denteado/metabolismo , MicroRNAs/metabolismo , Células-Tronco Neurais/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Animais , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Masculino , Ratos Sprague-Dawley
14.
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
15.
Pharmacology ; 103(3-4): 202-211, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30699432

RESUMO

AIM: The present study aimed to elucidate the effects of tramadol on the testicular functions of adult male rats due to the chronic usage of tramadol and the effect of its withdrawal. METHOD: Adult male albino rats were classified into the following 3 groups: (I) a control administered with normal saline and (II) tramadol-treated rats (40 mg/kg b.w. orally) for 21 successive days; and (III) like the rats in the second group but kept for 4 weeks after the last tramadol dose to study the effect of tramadol withdrawal. At the end of the experimental period, blood was collected and specimens from testis were taken for histopathological, biochemical, and molecular studies. A reverse transcription-polymerized chain reaction after RNA extraction from specimens was detected for the anti-apoptotic and pro-apoptotic genes in testicular tissues. Also, malondialdehyde (MDA) was measured in tissues homogenate and antioxidant enzymes activities were evaluated. RESULTS: The results of this study demonstrated histological changes in testicular tissues in groups II and III compared to the control group, accompanied with increased apoptotic index and proved by increased B-cell lymphoma-2 (Bcl-2) associated-X-protein and caspase-3 expression, whereas anti-apoptotic Bcl-2 markedly decreased. Moreover, in tramadol-abused and -withdrawal groups, the MDA level increased, while the antioxidant enzymes activity decreased and revealed oxidative stress, indicating that tramadol is harmful at the cellular level and can induce apoptotic changes in testicular tissues. The withdrawal effect showed signs of improvement, but it did not return to normal levels. CONCLUSIONS: It could be concluded that the administration of tramadol causes abnormalities on testicular tissues associated with oxidative stress, which confirmed the risk of increased oxidative stress on testicular tissues due to tramadol abuse.


Assuntos
Analgésicos Opioides , Apoptose , Transtornos Relacionados ao Uso de Opioides/patologia , Estresse Oxidativo , Espermatogênese , Síndrome de Abstinência a Substâncias/patologia , Testículo/fisiopatologia , Tramadol , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Modelos Animais de Doenças , Masculino , Transtornos Relacionados ao Uso de Opioides/metabolismo , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Ratos Wistar , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/fisiopatologia , Testículo/metabolismo , Testículo/patologia
16.
Addict Behav ; 92: 148-154, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30640146

RESUMO

BACKGROUND: Opioid dependence is a significant public health problem in the United States and the number of opioid overdose deaths among women has increased dramatically in comparison to men in the last few years. In this context, understanding the biological mechanisms underlying gender differences in vulnerability to opioid dependence is essential. METHODS: The current pilot study examined gender differences in subjective stress, heart rate (HR), and cortisol/dephydroepiandrosterone (DHEA) response to a laboratory stressor (Trier Social Stress Test; TSST) or a no-stress condition, and drug cue paradigm among men (n = 21) and women (n = 18) with opioid dependence. RESULTS: Significant group (TSST vs. no stress) differences emerged in self-reported stress [F(1,35) = 23.8, p < .001], HR [F(1,31) = 12.3; p = .001] and cortisol (F1,34 = 5.0; p = .032) response, such that the TSST group was more reactive than the no-stress group. Women reported greater subjective stress [F(1,35) = 6.5, p <= .015] in response to the TSST compared to men. However, men evidenced marginally greater cortisol and DHEA responses to the TSST compared to women [F(1,34) = 2.7; p = .113 and F(1,31) = 3.4; p = .073, respectively]. CONCLUSIONS: Although women with opioid dependence reported greater subjective stress when exposed to a laboratory stress paradigm as compared to men, the neuroendocrine response was more robust in men. This pattern was similar to gender findings in men and women with cocaine and tobacco use disorders. The blunted cortisol combined with an increased subjective response among women may be a sign of HPA axis dysregulation which could increase vulnerability to relapse in women.


Assuntos
Frequência Cardíaca/fisiologia , Hidrocortisona/metabolismo , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Transtornos Relacionados ao Uso de Opioides/metabolismo , Estresse Psicológico/epidemiologia , Estresse Psicológico/metabolismo , Adulto , Comorbidade , Feminino , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiopatologia , Masculino , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Projetos Piloto , Sistema Hipófise-Suprarrenal/metabolismo , Sistema Hipófise-Suprarrenal/fisiopatologia , Saliva/metabolismo , Fatores Sexuais , Estresse Psicológico/fisiopatologia
17.
ACS Chem Neurosci ; 9(10): 2288-2298, 2018 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-29851334

RESUMO

Opioid drug abuse and dependence/addiction are complex disorders regulated by a wide range of interacting networks of genes and pathways that control a variety of phenotypes. Although the field has been extensively progressed since the birth of the National Institute on Drug Abuse in 1974, the fundamental knowledge and involved mechanisms that lead to drug dependence/addiction are poorly understood, and thus, there has been limited success in the prevention of drug addiction and development of therapeutics for definitive treatment and cure of addiction disease. The lack of success in both identification of addiction in at-risk populations and the development of efficient drugs has resulted in a serious social and economic burden from opioid drug abuse with global increasing rate of mortality from drug overdoses. This perspective aims to draw the attention of scientists to the potential role of nanotechnologies, which might pave the way for the development of more practical platforms for either drug development or identification and screening of patients who may be vulnerable to addiction after using opioid drugs.


Assuntos
Encéfalo/metabolismo , Nanotecnologia , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Barreira Hematoencefálica , Encéfalo/diagnóstico por imagem , Meios de Contraste , Desenvolvimento de Medicamentos , Disbiose/metabolismo , Diagnóstico Precoce , Microbioma Gastrointestinal , Humanos , Programas de Rastreamento , Transtornos Relacionados ao Uso de Opioides/diagnóstico , Transtornos Relacionados ao Uso de Opioides/metabolismo , Terapêutica com RNAi , Medição de Risco , Detecção do Abuso de Substâncias , Transtornos Relacionados ao Uso de Substâncias/diagnóstico , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Transtornos Relacionados ao Uso de Substâncias/metabolismo
18.
Neurotoxicology ; 67: 245-258, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29906478

RESUMO

Nicotine and tramadol concomitant drug dependence pose increasing social, economic as well as public threats. Accordingly, the present study investigated neurochemical, neurobehavioral and neuropathological changes in the brain subsequent to the interaction of nicotine and tramadol. To this end, tramadol (20 mg/kg, i.p) and nicotine (0.25 mg/kg, i.p) were administrated to male albino mice once daily for 30 days. Consequent to microglial activation, nicotine exacerbated oxidative/nitrosative stress induced by tramadol as manifest by the step-up in thiobarbituric acid reactive substances and nitric oxide subsequent to the enhanced levels of neuronal and inducible nitric oxide synthases; paralleled by decreased non-protein sulfhydryls. Increased oxidative stress by tramadol and/or nicotine sequentially augmented nuclear factor kappa B and the proinflammatory cytokine tumor necrosis factor α with the induction of apoptosis evident by the increased caspase-3 immunoreactivity. However, paradoxical to the boosted inflammation and apoptosis, heightened DA levels in the cortex parallel along with increased tyrosine hydroxylase in midbrain were apparent. Concomitant administration of tramadol and nicotine impaired spatial navigation in the Morris Water Maze test coupled with enhanced levels of acetyl- and butyryl cholinestrases. However, tramadol in association with nicotine improved social interaction while decreasing anxiety and aggression linked to chronic administration of nicotine, effects manifested by increased levels of serotonin and GABA. These results provide evidence that co-administration of tramadol and nicotine may enhance reward and dependence while reducing anxiety and aggression linked to nicotine administration. However, such combination exacerbated neurotoxic effects and elicited negative effects regarding learning and memory.


Assuntos
Analgésicos Opioides/administração & dosagem , Encéfalo/metabolismo , Mediadores da Inflamação/metabolismo , Nicotina/administração & dosagem , Transtornos Relacionados ao Uso de Opioides/metabolismo , Tramadol/administração & dosagem , Analgésicos Opioides/toxicidade , Animais , Encéfalo/efeitos dos fármacos , Quimioterapia Combinada , Mediadores da Inflamação/antagonistas & inibidores , Relações Interpessoais , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Memória/efeitos dos fármacos , Memória/fisiologia , Camundongos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Agonistas Nicotínicos/administração & dosagem , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/psicologia , Tramadol/toxicidade
19.
Behav Brain Res ; 347: 368-376, 2018 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-29604365

RESUMO

Hydrocodone (HYD) is one of the most widely prescribed opioid analgesic drugs. Several neurotransmitters are involved in opioids relapse. Among these neurotransmitters, glutamate is suggested to be involved in opioid dependence and relapse. Glutamate is regulated by several glutamate transporters, including glutamate transporter 1 (GLT-1) and cystine/glutamate transporter (xCT). In this study, we investigated the effects of ceftriaxone (CEF) (200 mg/kg, i.p.), known to upregulate GLT-1 and xCT, on reinstatement to HYD (5 mg/kg, i.p.) using the conditioned place preference (CPP) paradigm in alcohol-preferring (P) rats. Animals were divided into three groups: 1) saline-saline group (SAL-SAL); 2) HYD-SAL group; and 3) HYD-CEF group. The CPP was conducted as follows: habituation phase, conditioning phase with HYD (i.p.) injections every other day for four sessions, extinction phase with CEF (i.p.) injections every other day for four sessions, and reinstatement phase with one priming dose of HYD. Time spent in the HYD-paired chamber after conditioning training was increased as compared to pre-conditioning. There was an increase in time spent in the HYD-paired chamber with one priming dose of HYD in the reinstatement test. HYD exposure downregulated xCT expression in the nucleus accumbens and hippocampus, but no effects were observed in the dorsomedial prefrontal cortex and amygdala. Importantly, CEF treatment attenuated the reinstatement effect of HYD and normalized xCT expression in the affected brain regions. These findings demonstrate that the attenuating effect of HYD reinstatement with CEF might be mediated through xCT.


Assuntos
Ceftriaxona/farmacologia , Fármacos do Sistema Nervoso Central/farmacologia , Comportamento de Procura de Droga/efeitos dos fármacos , Comportamento de Procura de Droga/fisiologia , Hidrocodona/farmacologia , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Transtornos Relacionados ao Uso de Álcool , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animais , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Predisposição Genética para Doença , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Neuroglia/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Ratos , Especificidade da Espécie
20.
Int Rev Neurobiol ; 136: 177-197, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29056151

RESUMO

Neuropeptide Y (NPY) is a neuromodulator that is widely expressed throughout the central nervous system (CNS) and which is cosecreted with classic neurotransmitters including GABA and glutamate. There is a long history of research implicating a role for NPY in modulating neurobiological responses to alcohol (ethanol) as well as other drugs of abuse. Both ethanol exposure and withdrawal from chronic ethanol have been shown to produce changes in NPY and NPY receptor protein levels and mRNA expression in the CNS. Importantly, manipulations of NPY Y1 and Y2 receptor signaling have been shown to alter ethanol consumption and self-administration in a brain region-specific manner, with Y1 receptor activation and Y2 receptor blockade in regions of the extended amygdala promoting robust reductions of ethanol intake. Similar observations have been made in studies examining neurobiological responses to nicotine, psychostimulants, and opioids. When taken together with observations of potential genetic linkage between the NPY system and the human alcohol abuse disorders, NPY represents a promising target for treating problematic alcohol and drug use, and in protecting individuals from relapse during abstinence.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Transtornos Relacionados ao Uso de Álcool/metabolismo , Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Neuropeptídeo Y/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Receptores de Neuropeptídeo Y/metabolismo , Tabagismo/metabolismo , Consumo de Bebidas Alcoólicas/genética , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA