Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 367
Filtrar
1.
Sci Rep ; 13(1): 16443, 2023 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-37777587

RESUMO

Neuroblastoma, the most common type of pediatric extracranial solid tumor, causes 10% of childhood cancer deaths. Despite intensive multimodal treatment, the outcomes of high-risk neuroblastoma remain poor. We urgently need to develop new therapies with safe long-term toxicity profiles for rapid testing in clinical trials. Drug repurposing is a promising approach to meet these needs. Here, we investigated disulfiram, a safe and successful chronic alcoholism treatment with known anticancer and epigenetic effects. Disulfiram efficiently induced cell cycle arrest and decreased the viability of six human neuroblastoma cell lines at half-maximal inhibitory concentrations up to 20 times lower than its peak clinical plasma level in patients treated for chronic alcoholism. Disulfiram shifted neuroblastoma transcriptome, decreasing MYCN levels and activating neuronal differentiation. Consistently, disulfiram significantly reduced the protein level of lysine acetyltransferase 2A (KAT2A), drastically reducing acetylation of its target residues on histone H3. To investigate disulfiram's anticancer effects in an in vivo model of high-risk neuroblastoma, we developed a disulfiram-loaded emulsion to deliver the highly liposoluble drug. Treatment with the emulsion significantly delayed neuroblastoma progression in mice. These results identify KAT2A as a novel target of disulfiram, which directly impacts neuroblastoma epigenetics and is a promising candidate for repurposing to treat pediatric neuroblastoma.


Assuntos
Dissulfiram , Neuroblastoma , Animais , Criança , Humanos , Camundongos , Dissuasores de Álcool/farmacologia , Dissuasores de Álcool/uso terapêutico , Linhagem Celular Tumoral , Dissulfiram/farmacologia , Dissulfiram/uso terapêutico , Regulação para Baixo , Reposicionamento de Medicamentos , Emulsões/uso terapêutico , Histona Acetiltransferases/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/genética
2.
JAMA ; 330(15): 1488-1490, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37707800

RESUMO

This study uses data from the 2013­March 2020 National Health and Nutrition Examination Survey to assess contemporary patterns of risky alcohol use among adults taking high-risk alcohol-interactive medications (benzodiazepine receptor agonists, opioids, and antiepileptics).


Assuntos
Dissuasores de Álcool , Consumo de Bebidas Alcoólicas , Interações Medicamentosas , Consumo de Bebidas Alcoólicas/epidemiologia , Assunção de Riscos , Comportamentos de Risco à Saúde , Humanos , Adulto , Estados Unidos/epidemiologia , Dissuasores de Álcool/classificação , Dissuasores de Álcool/farmacologia , Dissuasores de Álcool/uso terapêutico , Doença Crônica
3.
Eur J Pharmacol ; 926: 175035, 2022 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-35605658

RESUMO

As a key regulator involved in tumor development and progression, DJ-1 has been proposed as a potential therapeutic target against cancer. Also, the development of DJ-1 inhibitors holds great interests in cancer treatment. In the current study, by utilizing a small molecule covalent compounds library screening, we found that disulfiram (DSF), an FDA-approved chronic alcoholism drug, is a potent DJ-1 inhibitor. Glyoxalase assay and microscale thermophoresis analysis suggested that DSF exhibits strong inhibitory activity and high affinity to DJ-1 protein. Additionally, DSF similarly inhibited the methylglyoxal detoxification function of DJ-1 protein at the intracellular level. Notably, we discovered that DSF could significantly enhance N-(4-hydroxyphenyl) retinamide-based proliferation inhibition and apoptosis induction in different types of cancer cell lines, but not in normal tissue lines. Thus, our data suggest DSF functions as a potential inhibitor targeting DJ-1, which may provide a potential synergistic treatment option for cancer therapy.


Assuntos
Antineoplásicos , Dissulfiram , Neoplasias , Proteína Desglicase DJ-1 , Dissuasores de Álcool/farmacologia , Dissuasores de Álcool/uso terapêutico , Alcoolismo/tratamento farmacológico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Doença Crônica , Cobre , Dissulfiram/farmacologia , Dissulfiram/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Proteína Desglicase DJ-1/antagonistas & inibidores , Proteína Desglicase DJ-1/genética
4.
Drugs ; 82(3): 251-274, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35133639

RESUMO

Alcohol use disorder (AUD) is a highly prevalent but severely under-treated disorder, with only three widely-approved pharmacotherapies. Given that AUD is a very heterogeneous disorder, it is unlikely that one single medication will be effective for all individuals with an AUD. As such, there is a need to develop new, more effective, and diverse pharmacological treatment options for AUD with the hopes of increasing utilization and improving care. In this qualitative literature review, we discuss the efficacy, mechanism of action, and tolerability of approved, repurposed, and novel pharmacotherapies for the treatment of AUD with a clinical perspective. Pharmacotherapies discussed include: disulfiram, acamprosate, naltrexone, nalmefene, topiramate, gabapentin, varenicline, baclofen, sodium oxybate, aripiprazole, ondansetron, mifepristone, ibudilast, suvorexant, prazosin, doxazosin, N-acetylcysteine, GET73, ASP8062, ABT-436, PF-5190457, and cannabidiol. Overall, many repurposed and novel agents discussed in this review demonstrate clinical effectiveness and promise for the future of AUD treatment. Importantly, these medications also offer potential improvements towards the advancement of precision medicine and personalized treatment for the heterogeneous AUD population. However, there remains a great need to improve access to treatment, increase the menu of approved pharmacological treatments, and de-stigmatize and increase treatment-seeking for AUD.


Assuntos
Dissuasores de Álcool , Alcoolismo , Acamprosato , Dissuasores de Álcool/farmacologia , Dissuasores de Álcool/uso terapêutico , Alcoolismo/tratamento farmacológico , Baclofeno/uso terapêutico , Dissulfiram/farmacologia , Dissulfiram/uso terapêutico , Humanos , Naltrexona/uso terapêutico , Topiramato/uso terapêutico
5.
Pharmacol Res ; 174: 105923, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34607006

RESUMO

Idiopathic pulmonary fibrosis (IPF) induces significant morbidity and mortality, for which there are limited therapeutic options available. Here, we found that tetraethylthiuram disulphide (disulfiram, DSF), a derivative of thiuram, used in the treatment of alcohol abuse, has an inhibitory effect on bleomycin (BLM)-induced pulmonary fibrosis via the attenuation of the fibroblast-to-myofibroblast transition, migration, and proliferation of fibroblasts. Furthermore, DSF inhibited the activation of primary pulmonary fibroblasts and fibroblast cell line under transforming growth factor-ß 1 (TGF-ß1) challenge. Mechanistically, the anti-fibrotic effect of DSF on fibroblasts depends on the inhibition of TGF-ß signalling. We further determined that DSF interrupts the interaction between SMAD3 and TGF-ß receptor Ι (TBR Ι), and identified that DSF directly binds with SMAD3, in which Trp326, Thr330, and Cys332 of SMAD3 are critical binding sites for DSF. Collectively, our results reveal a powerful anti-fibrotic function of DSF in pulmonary fibrosis through the inhibition of TGF-ß/SMAD signalling in pulmonary fibroblasts, indicating that DSF is a promising therapeutic candidate for IPF.


Assuntos
Dissuasores de Álcool/uso terapêutico , Dissulfiram/uso terapêutico , Fibrose Pulmonar/tratamento farmacológico , Actinas/metabolismo , Dissuasores de Álcool/farmacologia , Animais , Bleomicina , Cadeia alfa 1 do Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I/metabolismo , Dissulfiram/farmacologia , Fibronectinas/genética , Fibronectinas/metabolismo , Células HEK293 , Humanos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
6.
PLoS Comput Biol ; 17(8): e1009110, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34351898

RESUMO

Ethanol is one of the most widely used recreational substances in the world and due to its ubiquitous use, ethanol abuse has been the cause of over 3.3 million deaths each year. In addition to its effects, ethanol's primary metabolite, acetaldehyde, is a carcinogen that can cause symptoms of facial flushing, headaches, and nausea. How strongly ethanol or acetaldehyde affects an individual depends highly on the genetic polymorphisms of certain genes. In particular, the genetic polymorphisms of mitochondrial aldehyde dehydrogenase, ALDH2, play a large role in the metabolism of acetaldehyde. Thus, it is important to characterize how genetic variations can lead to different exposures and responses to ethanol and acetaldehyde. While the pharmacokinetics of ethanol metabolism through alcohol dehydrogenase have been thoroughly explored in previous studies, in this paper, we combined a base physiologically-based pharmacokinetic (PBPK) model with a whole-body genome-scale model (WBM) to gain further insight into the effect of other less explored processes and genetic variations on ethanol metabolism. This combined model was fit to clinical data and used to show the effect of alcohol concentrations, organ damage, ALDH2 enzyme polymorphisms, and ALDH2-inhibiting drug disulfiram on ethanol and acetaldehyde exposure. Through estimating the reaction rates of auxiliary processes with dynamic Flux Balance Analysis, The PBPK-WBM was able to navigate around a lack of kinetic constants traditionally associated with PK modelling and demonstrate the compensatory effects of the body in response to decreased liver enzyme expression. Additionally, the model demonstrated that acetaldehyde exposure increased with higher dosages of disulfiram and decreased ALDH2 efficiency, and that moderate consumption rates of ethanol could lead to unexpected accumulations in acetaldehyde. This modelling framework combines the comprehensive steady-state analyses from genome-scale models with the dynamics of traditional PK models to create a highly personalized form of PBPK modelling that can push the boundaries of precision medicine.


Assuntos
Acetaldeído/metabolismo , Alcoolismo/genética , Alcoolismo/metabolismo , Etanol/metabolismo , Modelos Biológicos , Acetaldeído/farmacocinética , Acetaldeído/toxicidade , Inibidores de Acetaldeído Desidrogenases/farmacologia , Dissuasores de Álcool/farmacologia , Alcoolismo/tratamento farmacológico , Aldeído-Desidrogenase Mitocondrial/genética , Aldeído-Desidrogenase Mitocondrial/metabolismo , Biologia Computacional , Simulação por Computador , Dissulfiram/farmacologia , Etanol/farmacocinética , Etanol/toxicidade , Humanos , Absorção Intestinal/fisiologia , Cinética , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Distribuição Tecidual
7.
Toxicol Appl Pharmacol ; 426: 115642, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34242567

RESUMO

Disulfiram (DSF), a sulfur-containing compound, has been used to treat chronic alcoholism and cancer for decades by inactivating aldehyde dehydrogenase (ALDH). Hydrogen sulfide (H2S) is a new gasotransmitter and regulates various cellular functions by S-sulfhydrating cysteine in the target proteins. H2S exhibits similar properties to DSF in the sensitization of cancer cells. The interaction of DSF and H2S on ALDH activity and liver cancer cell survival are not clear. Here it was demonstrated that DSF facilitated H2S release from thiol-containing compounds, and DSF and H2S were both capable of regulating ALDH through inhibition of gene expression and enzymatic activity. The supplement of H2S sensitized human liver cancer cells (HepG2) to DSF-inhibited cell viability. The expression of cystathionine gamma-lyase (a major H2S-generating enzyme) was lower but ALDH was higher in mouse liver cancer stem cells (Dt81Hepa1-6) in comparison with their parental cells (Hepa1-6), and H2S was able to inhibit liver cancer stem cell adhesion. In conclusion, these data point to the potential of combining DSF and H2S for inhibition of cancer cell growth and tumor development by targeting ALDH.


Assuntos
Inibidores de Acetaldeído Desidrogenases/farmacologia , Dissuasores de Álcool/farmacologia , Aldeído Desidrogenase/antagonistas & inibidores , Antineoplásicos/farmacologia , Dissulfiram/farmacologia , Sulfeto de Hidrogênio/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Aldeído Desidrogenase/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cobre/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Fígado/efeitos dos fármacos , Fígado/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Camundongos , Temperatura
8.
Biophys Chem ; 276: 106610, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34089978

RESUMO

In the new millennium, the outbreak of new coronavirus has happened three times: SARS-CoV, MERS-CoV, and SARS-CoV-2. Unfortunately, we still have no pharmaceutical weapons against the diseases caused by these viruses. The pandemic of SARS-CoV-2 reminds us the urgency to search new drugs with totally different mechanism that may target the weaknesses specific to coronaviruses. Herein, we disclose a computational evaluation of targeted oxidation strategy (TOS) for potential inhibition of SARS-CoV-2 by disulfiram, a 70-year-old anti-alcoholism drug, and predict a multiple-target mechanism. A preliminary list of promising TOS drug candidates targeting the two thiol proteases of SARS-CoV-2 are proposed upon virtual screening of 32,143 disulfides.


Assuntos
Dissuasores de Álcool/química , Antivirais/química , Proteases 3C de Coronavírus/antagonistas & inibidores , Proteases Semelhantes à Papaína de Coronavírus/antagonistas & inibidores , Dissulfiram/química , Inibidores de Proteases/química , SARS-CoV-2/química , Dissuasores de Álcool/farmacologia , Antivirais/farmacologia , Domínio Catalítico , Proteases 3C de Coronavírus/química , Proteases 3C de Coronavírus/genética , Proteases 3C de Coronavírus/metabolismo , Proteases Semelhantes à Papaína de Coronavírus/química , Proteases Semelhantes à Papaína de Coronavírus/genética , Proteases Semelhantes à Papaína de Coronavírus/metabolismo , Dissulfiram/farmacologia , Reposicionamento de Medicamentos , Expressão Gênica , Humanos , Cinética , Simulação de Acoplamento Molecular , Oxirredução , Inibidores de Proteases/farmacologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Teoria Quântica , SARS-CoV-2/enzimologia , Especificidade por Substrato , Termodinâmica , Tratamento Farmacológico da COVID-19
9.
Addict Biol ; 26(2): e12908, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32329567

RESUMO

The prevalence of alcohol use disorders (AUDs) has steadily increased in the United States over the last 30 years. Alcohol acts on multiple receptor systems including the nicotinic acetylcholine receptors (nAChRs), which are known to mediate alcohol consumption and reward. We previously reported that the preclinical drug sazetidine-A, a nAChR agonist and desensitizer, reduces alcohol consumption without affecting nicotine consumption in C57BL/6J mice. Here, we found that sazetidine-A enhances the expression of alcohol aversion without affecting the expression or acquisition of conditioned alcohol reward in C57BL/6J mice. Microinjection of sazetidine-A into the ventral midbrain targeting the ventral tegmental area (VTA) reduced binge alcohol consumption, implicating this region in mediating the effects of sazetidine-A. Furthermore, the sazetidine-A-induced reduction in alcohol consumption was mediated by non-α4 containing nAChRs, as sazetidine-A reduced binge alcohol consumption in both α4 knock-out and wild-type mice. Finally, we found that in mice pretreated with sazetidine-A, alcohol induced Fos transcript in Th-, but not Gad2-expressing neurons in the VTA as measured by increased Fos transcript expression. In summary, we find that sazetidine-A enhances the expression of alcohol aversion, which may underlie the reduction in alcohol consumption induced by sazetidine-A. Elucidating the identity of non-α4 nAChRs in alcohol aversion mechanisms will provide a better understanding the complex role of nAChRs in alcohol addiction and potentially reveal novel drug targets to treat AUDs.


Assuntos
Dissuasores de Álcool/farmacologia , Alcoolismo/tratamento farmacológico , Azetidinas/farmacologia , Agonistas Nicotínicos/farmacologia , Piridinas/farmacologia , Animais , Consumo Excessivo de Bebidas Alcoólicas/tratamento farmacológico , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Receptores Nicotínicos , Recompensa , Área Tegmentar Ventral/efeitos dos fármacos
10.
Alcohol Clin Exp Res ; 44(10): 2118-2129, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33043520

RESUMO

BACKGROUND: The misuse and abuse of alcohol is a major public health issue. However, available treatments are limited with variable efficacy. Recently, preclinical studies show that glucagon-like-peptide-1 (GLP-1) and its analogue Exendin-4 (Ex4) potently reduce a range of alcohol intake behaviors, thus highlighting its potential as a treatment for alcohol use disorders. However, the neural mechanisms and sites of action mediating the effects of Ex4 on alcohol intake behaviors remain to be characterized. This study examined the ventral tegmental area (VTA) as a site of action for the effects of GLP-1 on alcohol intake. METHODS: Male Long-Evans rats were given intermittent access to 20% alcohol and trained to nose poke for 20% alcohol. Rats received intra-VTA injections of Ex4 (vehicle, 0.01, 0.05 µg), and the effects of VTA Ex4 on alcohol self-administration, motivation, and relapse were assessed. RESULTS: When compared to vehicle treatment, intra-VTA Ex4 (0.01, 0.05 µg) delivery significantly reduced alcohol self-administration, an effect that was particularly prominent in high alcohol drinkers. However, VTA Ex4 did not reduce reacquisition of alcohol self-administration after extinction nor the motivation to obtain alcohol. Importantly, the lower dose of Ex4 (0.01 µg) used had no effect on food intake or locomotor activity, suggesting that the reduction in alcohol self-administration observed was not secondary to caloric intake or motor deficits. CONCLUSIONS: Together, these findings provide support for the VTA as a key site of action for GLP-1 on alcohol self-administration but not the reacquisition of alcohol self-administration or motivation to work for alcohol.


Assuntos
Dissuasores de Álcool/farmacologia , Consumo de Bebidas Alcoólicas/tratamento farmacológico , Exenatida/farmacologia , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Dissuasores de Álcool/metabolismo , Dissuasores de Álcool/uso terapêutico , Consumo de Bebidas Alcoólicas/metabolismo , Consumo de Bebidas Alcoólicas/fisiopatologia , Animais , Exenatida/metabolismo , Exenatida/uso terapêutico , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/fisiologia , Masculino , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Long-Evans , Área Tegmentar Ventral/metabolismo , Área Tegmentar Ventral/fisiopatologia
11.
Drug Alcohol Depend ; 217: 108272, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32971391

RESUMO

BACKGROUND: For people with HIV (PWH) and alcohol use disorder (AUD) who initiated behavioral treatment (BAUD) we: 1) describe BAUD intensity and medication (MAUD); and 2) examine whether BAUD and MAUD were associated with changes in HIV-related outcomes (CD4 cell count, HIV-1 viral load [VL], VACS Index score 2.0, and antiretroviral [ARV] adherence) from before to one year after treatment initiation. METHODS: We used Veterans Aging Cohort Study (VACS) data to describe BAUD intensity and MAUD (acamprosate, disulfiram, and naltrexone, gabapentin or topiramate). Linear regression models estimated changes in outcomes and included BAUD, MAUD, age and race/ethnicity. RESULTS: We identified 7830 PWH who initiated BAUD from 01/2008-09/2017. Median age was 53, 60% were African-American and 28% white. BAUD intensity groups were: 1) Single Visit - 35%; 2) Minimal - 44% recieved ∼2 visits during first month; 3) Sustained Moderate - 17% recieved ∼8 visits/month initially; and 4) Intensive - 4% started out receiving ∼14-16 visits/month. Only 9% recieved MAUD, the majority of which was gabapentin. Among those with detectable VL: all HIV-related outcomes improved more among those with more intensive BAUD. Among those with undetectable VL: adherence improved more among those with greater BAUD intensity. MAUD was associated with increased CD4 among those with detectable VL and with improved adherence among both groups. CONCLUSION: Of those with >1 BAUD visit, only 21% received at least moderate BAUD and 9% received at least 6 months of MAUD. Increasing AUD treatment intensity may improve HIV-related outcomes, especially among those with detectable VL.


Assuntos
Alcoolismo/tratamento farmacológico , Alcoolismo/psicologia , Terapia Cognitivo-Comportamental/métodos , Infecções por HIV/tratamento farmacológico , Infecções por HIV/psicologia , Veteranos/psicologia , Adulto , Dissuasores de Álcool/farmacologia , Dissuasores de Álcool/uso terapêutico , Alcoolismo/epidemiologia , Antirretrovirais/farmacologia , Antirretrovirais/uso terapêutico , Terapia Cognitivo-Comportamental/tendências , Estudos de Coortes , Feminino , Infecções por HIV/epidemiologia , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Carga Viral/efeitos dos fármacos , Carga Viral/tendências
12.
Alcohol Clin Exp Res ; 44(6): 1224-1233, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32406553

RESUMO

BACKGROUND: Human laboratory paradigms are a pillar in medication development for alcohol use disorders (AUD). Neuroimaging paradigms, in which individuals are exposed to cues that elicit neural correlates of alcohol craving (e.g., mesocorticolimbic activation), are increasingly utilized to test the effects of AUD medications. Elucidation of the translational effects of these neuroimaging paradigms on human laboratory paradigms, such as self-administration, is warranted. The current study is a secondary analysis examining whether alcohol cue-induced activation in the ventral striatum is predictive of subsequent alcohol self-administration in the laboratory. METHODS: Non-treatment-seeking heavy drinkers of East Asian descent (n = 41) completed a randomized, placebo-controlled, double-blind, crossover experiment on the effects of naltrexone on neuroimaging and human laboratory paradigms. Participants completed 5 days of study medication (or placebo); on day 4, they completed a neuroimaging alcohol taste cue-reactivity task. On the following day (day 5), participants completed a 60-minute alcohol self-administration paradigm. RESULTS: Multilevel Cox regressions indicated a significant effect of taste cue-elicited ventral striatum activation on latency to first drink, Wald χ2  = 2.88, p = 0.05, such that those with higher ventral striatum activation exhibited shorter latencies to consume their first drink. Similarly, ventral striatum activation was positively associated with total number of drinks consumed, F(1, 38) = 5.90, p = 0.02. These effects were significant after controlling for alcohol use severity, OPRM1 genotype, and medication. Other potential regions of interest (anterior cingulate, thalamus) were not predictive of self-administration outcomes. CONCLUSIONS: Neuroimaging alcohol taste cue paradigms may be predictive of laboratory paradigms such as self-administration. Elucidation of the relationships among different paradigms will inform how these paradigms may be used synergistically in experimental medicine and medication development.


Assuntos
Transtornos Relacionados ao Uso de Álcool/diagnóstico por imagem , Depressores do Sistema Nervoso Central/administração & dosagem , Sinais (Psicologia) , Etanol/administração & dosagem , Estriado Ventral/diagnóstico por imagem , Adulto , Dissuasores de Álcool/farmacologia , Transtornos Relacionados ao Uso de Álcool/fisiopatologia , Família Aldeído Desidrogenase 1/genética , Aldeído-Desidrogenase Mitocondrial/genética , Feminino , Neuroimagem Funcional , Genótipo , Giro do Cíngulo/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética , Masculino , Análise Multinível , Naltrexona/farmacologia , Modelos de Riscos Proporcionais , Distribuição Aleatória , Receptores Opioides mu/genética , Autoadministração , Tálamo/diagnóstico por imagem , Estriado Ventral/efeitos dos fármacos , Estriado Ventral/fisiopatologia , Adulto Jovem
13.
J Nepal Health Res Counc ; 18(1): 75-81, 2020 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-32335597

RESUMO

BACKGROUND: Relapse prevention in alcoholism is recognised as an important component of management. Use of pharmacotherapies to prevent relapse in combination to psychological intervention is emerging. Disulfiram and Naltrexone are two of three FDA approved drugs for pharmacotherapy. The aim of the study is to compare the effectiveness of these two drugs in preventing relapse in alcohol dependence syndrome cases. METHODS: A prospective crossectional study was conducted to compare disulfiram and naltrexone in alcohol dependent patients in tertiary institution. Cases of alcohol dependence syndrome were diagnosed based on ICD-10 DCR presenting to psychiatry department of Tribhuvan University Teaching Hospital, over the period of 6 months. After detoxification and fulfillment of inclusion criteria, semi structured proforma, Severity of alcohol dependence questionnaire, Stages of change readiness and treatment eagerness scale, Obsessive compulsive drinking scale were applied. Drug allocation was based on simple random method and on subsequent follow ups done at 2nd, 4th, 8th, 12th week semi structured proforma, Obsessive Compulsive Drinking Scale were completed and psychological intervention continued. After data collection, analysis and final results were computed. RESULTS: Both drugs reduced craving and amount of alcohol intake(p less than 0.001). Relapse was more in naltrexone group but was not statistically significant (p>0.05). Side effects were more with disulfiram(p less than 0.001) whereas dropout was more in naltrexone group,(p less than 0.01). CONCLUSIONS: Disulfiram and Naltrexone were equally effective in reducing craving, reducing amount of alcohol intake, and preventing relapse in 12 weeks follow up period. Naltrexone was found to be better in tolerability whereas disulfiram was better in terms of dropout from treatment.


Assuntos
Dissuasores de Álcool/farmacologia , Alcoolismo/tratamento farmacológico , Dissulfiram/farmacologia , Naltrexona/farmacologia , Adulto , Dissuasores de Álcool/efeitos adversos , Estudos Transversais , Dissulfiram/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Naltrexona/efeitos adversos , Nepal , Estudos Prospectivos , Recidiva , Resultado do Tratamento
14.
Cells ; 9(2)2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32085572

RESUMO

Research on repurposing the old alcohol-aversion drug disulfiram (DSF) for cancer treatment has identified inhibition of NPL4, an adaptor of the p97/VCP segregase essential for turnover of proteins involved in multiple pathways, as an unsuspected cancer cell vulnerability. While we reported that NPL4 is targeted by the anticancer metabolite of DSF, the bis-diethyldithiocarbamate-copper complex (CuET), the exact, apparently multifaceted mechanism(s) through which the CuET-induced aggregation of NPL4 kills cancer cells remains to be fully elucidated. Given the pronounced sensitivity to CuET in tumor cell lines lacking the genome integrity caretaker proteins BRCA1 and BRCA2, here we investigated the impact of NPL4 targeting by CuET on DNA replication dynamics and DNA damage response pathways in human cancer cell models. Our results show that CuET treatment interferes with DNA replication, slows down replication fork progression and causes accumulation of single-stranded DNA (ssDNA). Such a replication stress (RS) scenario is associated with DNA damage, preferentially in the S phase, and activates the homologous recombination (HR) DNA repair pathway. At the same time, we find that cellular responses to the CuET-triggered RS are seriously impaired due to concomitant malfunction of the ATRIP-ATR-CHK1 signaling pathway that reflects an unorthodox checkpoint silencing mode through ATR (Ataxia telangiectasia and Rad3 related) kinase sequestration within the CuET-evoked NPL4 protein aggregates.


Assuntos
Dissuasores de Álcool/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Dano ao DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Dissulfiram/farmacologia , Neoplasias/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem/metabolismo , Proteínas de Ligação a DNA/metabolismo , Humanos , Neoplasias/patologia , Proteínas Nucleares/metabolismo , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas/induzido quimicamente , Transdução de Sinais/efeitos dos fármacos , Proteína com Valosina/metabolismo
15.
Psychol Addict Behav ; 34(1): 10-22, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31750701

RESUMO

Resurgence may be a mechanism of relapse in alcohol use disorder patients upon discharge from treatment as part of an abuse-treatment-relapse cycle. Adjunctive pharmacotherapies may be a means to facilitate behavioral treatments and block resurgence. Experiments were conducted using a model of alcohol self-administration to assess the repeatability of the elimination and resurgence of alcohol-maintained behavior and the effects of naltrexone. Experiments had three phases. In Phase 1, behavior was maintained by oral alcohol under a fixed-ratio schedule. In Phase 2, behavior was extinguished via condensed milk delivery under a differential-reinforcement-of-other-behavior (DRO) schedule. In Phase 3, the DRO schedule was eliminated. In Experiment 1, this 3-phase cycle was replicated 4 times. Across replications, response rates and dose of alcohol consumed did not differ in Phase 1, alcohol-maintained behavior was eliminated more rapidly in Phase 2, and the resurgence effect was generally stable in Phase 3. In Experiment 2, naltrexone was administered in Phase 2, Phase 3, or both Phases 2 and 3, to separate groups of rats. Naltrexone facilitated the elimination of alcohol-maintained behavior in Phase 2 and, the resurgence of alcohol-maintained behavior was reduced only for those rats that received naltrexone in both phases. Together, these experiments demonstrate that the resurgence of alcohol-maintained behavior is replicable within-subjects and, further, resurgence of alcohol-maintained behavior may be a useful model to evaluate pharmacological interventions to facilitate behavioral treatments and reduce the likelihood of relapse. Results with naltrexone support the use of medication-assisted therapy approaches to reduce relapse risk in patients. (PsycINFO Database Record (c) 2020 APA, all rights reserved).


Assuntos
Dissuasores de Álcool/farmacologia , Alcoolismo , Condicionamento Operante/efeitos dos fármacos , Etanol/administração & dosagem , Extinção Psicológica/efeitos dos fármacos , Naltrexona/farmacologia , Reforço Psicológico , Animais , Masculino , Ratos , Autoadministração
16.
Eur Neuropsychopharmacol ; 29(12): 1386-1396, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31679889

RESUMO

The mechanisms behind relapse to ethanol intake in recovering alcoholics are still unclear. The negative reinforcing effects contributing to ethanol addiction, including relapse, are considered to be partly driven by the κ-opioidergic system. As the κ-opioidergic system interacts with the mesolimbic reward pathway, the aim of the study was to clarify the role of nucleus accumbens shell κ-opioidergic mechanisms in relapse to ethanol intake by using the alcohol deprivation effect (ADE) paradigm. The ADE is defined as a transient increase in voluntary ethanol intake after a forced period of abstinence. Male Long-Evans rats were trained to voluntarily consume 10% (v/v) ethanol solution. Ethanol access and deprivation cycles were initiated after stable ethanol intake baselines had been reached and bilateral guide cannulas had been implanted above the nucleus accumbens shell. One cycle consisted of 10 days of 90 min access to ethanol followed by 6 days of ethanol deprivation. The ADE was measured in the beginning of a new cycle. Rats received JDTic, a selective κ-antagonist, either subcutaneously (10 mg/kg) or intra-accumbally (15 µg/site) or, as a reference substance, systemic naltrexone (0.3 mg/kg) before ethanol re-access, and the effects on the ADE were evaluated. Systemic and intra-accumbal JDTic significantly attenuated the ADE on the first day of ethanol re-access, as did systemic naltrexone. Additionally, naltrexone decreased ethanol intake levels. These results suggest that nucleus accumbens shell κ-opioidergic mechanisms may have a role in mediating relapse to ethanol intake. Additionally, κ-antagonism could be a valuable adjunct in ethanol relapse prevention.


Assuntos
Abstinência de Álcool , Consumo de Bebidas Alcoólicas/prevenção & controle , Antagonistas de Entorpecentes/uso terapêutico , Piperidinas/uso terapêutico , Receptores Opioides kappa/antagonistas & inibidores , Tetra-Hidroisoquinolinas/uso terapêutico , Abstinência de Álcool/psicologia , Dissuasores de Álcool/farmacologia , Dissuasores de Álcool/uso terapêutico , Consumo de Bebidas Alcoólicas/metabolismo , Consumo de Bebidas Alcoólicas/psicologia , Animais , Etanol/administração & dosagem , Masculino , Naltrexona/farmacologia , Naltrexona/uso terapêutico , Antagonistas de Entorpecentes/farmacologia , Piperidinas/farmacologia , Ratos , Ratos Long-Evans , Receptores Opioides kappa/metabolismo , Autoadministração , Tetra-Hidroisoquinolinas/farmacologia
17.
J Subst Abuse Treat ; 104: 7-14, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31370987

RESUMO

Medication-assisted behavior treatment for alcohol use disorder (AUD) holds promise to enhance the efficacy of medication and of behavior therapy when administered individually. The present study examines the treatment benefit of combined outpatient naltrexone (NTX) treatment with Alcoholics Anonymous Facilitation (AAF) behavior therapy, in the context of OPRM1 genotype. The minor OPRM1 Asp40 G-allele has been associated with greater positive reinforcing effects of alcohol consumption and greater alcohol craving, suggesting that individuals carrying the OPRM1 G allele may have an improved naltrexone response. Twenty patients, including 7 G-allele carriers, received 90 days of naltrexone with medication support and dispensing sessions, and ten AAF behavior therapy sessions. During treatment and the eight-week posttreatment follow-up, an overall increase in percent days abstinent was observed for the sample as a whole, but G-allele carriers reported relatively heavier drinking relative to other subjects. These findings suggest that this enhanced medication-assisted behavior treatment is a promising therapeutic combination, and mirror other recent findings that G-allele carriers may require more intensive treatment.


Assuntos
Dissuasores de Álcool/farmacologia , Alcoolismo/genética , Alcoolismo/terapia , Terapia Comportamental , Naltrexona/farmacologia , Avaliação de Resultados em Cuidados de Saúde , Receptores Opioides mu/genética , Adulto , Alcoolismo/tratamento farmacológico , Terapia Combinada , Feminino , Variação Genética , Humanos , Masculino , Pessoa de Meia-Idade
18.
J Toxicol Sci ; 44(8): 535-542, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31378764

RESUMO

Spinocerebellar ataxia type 3 (SCA3) is caused by the expansion of a glutamine-encoding CAG repeat in the ATXN3 gene encoding the protein ataxin-3. The nuclear presence of polyglutamine-expanded ataxin-3 is of critical importance for the pathogenesis of SCA3. Disulfiram, an FDA-approved drug for alcoholism, has also garnered attention in cancer treatment. However, it has shown toxicity in the nervous system. Bearing this in mind, we treated cells expressing ataxin-3 with disulfiram to measure several pathogenic cascades of SCA3, including aggregate formation, soluble ataxin-3 expression and nuclear localization of ataxin-3 and the cytotoxicity, which assess the direct effect of disulfiram on SCA3 cell models. To our knowledge, this is direct evidence that disulfiram elevated the nuclear localization of polyglutamine-expanded ataxin-3 and enhanced the cytotoxicity in a cell model of SCA3. Furthermore, disulfiram did not affect the aggregate formation of polyglutamine-expanded ataxin-3 at least at a single dose. Our findings repurpose disulfiram as a modulator of ataxin-3 nuclear transport that aggravates the pathology of SCA3, which is a new target for disulfiram. This study also represents an important example of determining novel side effects in pre-existing drugs. This study suggests that caution may be warranted when this compound is used to treat alcohol abuse or cancer in patients carrying a SCA3-causing mutation.


Assuntos
Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Dissuasores de Álcool/farmacologia , Dissuasores de Álcool/toxicidade , Ataxina-3/metabolismo , Dissulfiram/farmacologia , Dissulfiram/toxicidade , Doença de Machado-Joseph/etiologia , Dissuasores de Álcool/efeitos adversos , Dissulfiram/efeitos adversos , Células HEK293 , Humanos , Doença de Machado-Joseph/genética
19.
Addict Biol ; 24(6): 1138-1152, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31148304

RESUMO

Naltrexone (NTX) has been widely studied for the treatment of alcohol use disorder with overall support for its efficacy. The mechanisms of action of naltrexone are thought to involve attenuation of the hedonic effects of alcohol and potentiation of its aversive effects. In order to provide a quantitative estimate of the effects of naltrexone on subjective response to alcohol, the aims of this meta-analytic review are to examine the effects of naltrexone across four domains of subjective response. Meta-analyses of naltrexone effects on alcohol craving (k = 16, N = 686), stimulation (k = 15, N = 675), sedation (k = 18, N = 777), and negative mood (k = 9, N = 281) suggested that under laboratory conditions and compared with placebo, naltrexone reduces craving (Hedges g = -0.252; SE = 0.054; 95% CI, -0.375 to -0.130; P < 0.01), reduces stimulation (g = -0.223; SE = 0.067; 95% CI, -0.372 to -0.074; P < 0.01), increases sedation (g = 0.251; SE = 0.064; 95% CI, 0.112-0.389; P < 0.01), and increases negative mood (g = 0.227; SE = 0.047; 95% CI, 0.100-0.354; P < 0.01). Results were robust when drinks per month and alcohol dose were added to the models as covariates. The effects of naltrexone varied by severity of alcohol use with medication effects on craving and stimulation being observed in sample of both heavy drinkers and AUD individuals. These results are consistent with the hypothesized mechanisms of action of NTX, although the effects are of small magnitude. This meta-analysis aggregates across multiple human laboratory studies of NTX's effects on subjective response to alcohol, providing a comprehensive summary of a key mechanism of NTX efficacy, namely, alteration of the subjective experience of alcohol.


Assuntos
Afeto/efeitos dos fármacos , Dissuasores de Álcool/farmacologia , Depressores do Sistema Nervoso Central/farmacologia , Fissura/efeitos dos fármacos , Etanol/farmacologia , Naltrexona/farmacologia , Alcoolismo/tratamento farmacológico , Humanos , Hipnóticos e Sedativos
20.
Neurosci Biobehav Rev ; 103: 384-398, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31112713

RESUMO

Alcohol Use Disorder (AUD) is a chronic disease that develops over the years. The complexity of the neurobiological processes contributing to the emergence of AUD and the neuroadaptive changes occurring during disease progression make it difficult to improve treatments. On the other hand, this complexity offers researchers the possibility to explore new targets. Over years of intense research several molecules were tested in AUD; in most cases, despite promising preclinical data, the clinical efficacy appeared insufficient to justify futher development. A prototypical example is that of corticotropin releasing factor type 1 receptor (CRF1R) antagonists that showed significant effectiveness in animal models of AUD but were largely ineffective in humans. The present article attempts to analyze the most recent venues in the development of new medications in AUD with a focus on the most promising drug targets under current exploration. Moreover, we delineate the importance of using a more integrated translational framework approach to correlate preclinical findings and early clinical data to enhance the probability to validate biological targets of interest.


Assuntos
Dissuasores de Álcool/farmacologia , Alcoolismo/tratamento farmacológico , Alcoolismo/metabolismo , Anticonvulsivantes/farmacologia , Agonistas dos Receptores de GABA-B/farmacologia , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Alcoolismo/diagnóstico por imagem , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...