Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 268
Filtrar
1.
medRxiv ; 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38746260

RESUMEN

Background: The prevalence of co-occurring heavy alcohol consumption and obesity is increasing in the United States. Despite neurobiological overlap in the regulation of alcohol consumption and eating behavior, alcohol- and body mass index (BMI)-related phenotypes show no or minimal genetic correlation. We hypothesized that the lack of genetic correlation is due to mixed effect directions of variants shared by AUD and BMI. Methods: We applied MiXeR, to investigate shared genetic architecture between AUD and BMI in individuals of European ancestry. We used conjunctional false discovery rate (conjFDR) analysis to detect loci associated with both phenotypes and their directional effect, Functional Mapping and Annotation (FUMA) to identify lead single nucleotide polymorphisms (SNPs), Genotype-Tissue Expression (GTEx) samples to examine gene expression enrichment across tissue types, and BrainXcan to evaluate the shared associations of AUD and BMI with brain image-derived phenotypes. Results: MiXeR analysis indicated polygenic overlap of 80.9% between AUD and BMI, despite a genetic correlation (r g ) of -.03. ConjFDR analysis yielded 56 lead SNPs with the same effect direction and 76 with the opposite direction. Of the 132 shared lead SNPs, 53 were novel for both AUD and BMI. GTEx analyses identified significant overexpression in the frontal cortex (BA9), hypothalamus, cortex, anterior cingulate cortex (BA24), hippocampus, and amygdala. Amygdala and caudate nucleus gray matter volumes were significantly associated with both AUD and BMI in BrainXcan analyses. Conclusions: More than half of variants significantly associated with AUD and BMI had opposite directions of effect for the traits, supporting our hypothesis that this is the basis for their lack of genetic correlation. Follow-up analyses identified brain regions implicated in executive functioning, reward, homeostasis, and food intake regulation. Together, these findings clarify the extensive polygenic overlap between AUD and BMI and elucidate several overlapping neurobiological mechanisms.

2.
Biomed Pharmacother ; 174: 116595, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38640709

RESUMEN

Fatty liver is the earliest response of the liver to excessive alcohol consumption. Previously we identified that chronic alcohol administration increases levels of stomach-derived hormone, ghrelin, which by reducing circulating insulin levels, ultimately contributes to the development of alcohol-associated liver disease (ALD). In addition, ghrelin directly promotes fat accumulation in hepatocytes by enhancing de novo lipogenesis. Other than promoting ALD, ghrelin is known to increase alcohol craving and intake. In this study, we used a ghrelin receptor (GHSR) knockout (KO) rat model to characterize the specific contribution of ghrelin in the development of ALD with emphasis on energy homeostasis. Male Wistar wild type (WT) and GHSR-KO rats were pair-fed the Lieber-DeCarli control or ethanol diet for 6 weeks. At the end of the feeding period, glucose tolerance test was conducted, and tissue samples were collected. We observed reduced alcohol intake by GHSR-KOs compared to a previous study where WT rats were fed ethanol diet ad libitum. Further, when the WTs were pair-fed to GHSR-KOs, the KO rats exhibited resistance to develop ALD through improving insulin secretion/sensitivity to reduce adipose lipolysis and hepatic fatty acid uptake/synthesis and increase fatty acid oxidation. Furthermore, proteomic data revealed that ethanol-fed KO exhibit less alcohol-induced mitochondrial dysfunction and oxidative stress than WT rats. Proteomic data also confirmed that the ethanol-fed KOs are insulin sensitive and are resistant to hepatic steatosis development compared to WT rats. Together, these data confirm that inhibiting ghrelin action prevent alcohol-induced liver and adipose dysfunction independent of reducing alcohol intake.


Asunto(s)
Etanol , Ghrelina , Hepatopatías Alcohólicas , Hígado , Ratas Wistar , Receptores de Ghrelina , Animales , Masculino , Ratas , Consumo de Bebidas Alcohólicas , Ácidos Grasos/metabolismo , Ghrelina/metabolismo , Insulina/metabolismo , Insulina/sangre , Resistencia a la Insulina , Hígado/metabolismo , Hígado/efectos de los fármacos , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/patología , Estrés Oxidativo/efectos de los fármacos , Proteómica/métodos , Receptores de Ghrelina/metabolismo , Receptores de Ghrelina/genética
4.
ACS Chem Neurosci ; 15(8): 1609-1610, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38551493

RESUMEN

Amylin is a neuroendocrine hormone with a potential role in addictive disorders, including alcohol use disorder (AUD). In addition to reducing appetitive behavior, amylin has been shown to affect alcohol-related behaviors in rodents. Delineating the biobehavioral correlates of amylin in relation to alcohol seeking and consumption has the potential of identifying new treatment targets for AUD, yet additional translational and human research is needed.


Asunto(s)
Alcoholismo , Conducta Adictiva , Hormonas Peptídicas , Humanos , Alcoholismo/tratamiento farmacológico , Polipéptido Amiloide de los Islotes Pancreáticos , Etanol
5.
Neuropsychopharmacology ; 49(6): 1042-1049, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38409282

RESUMEN

The stomach-derived hormone ghrelin plays not only a role in feeding, starvation, and survival, but it has been suggested to also be involved in the stress response, in neuropsychiatric conditions, and in alcohol and drug use disorders. Mechanisms related to reward processing might mediate ghrelin's broader effects on complex behaviors, as indicated by animal studies and mostly correlative human studies. Here, using a within-subject double-blind placebo-controlled design with intravenous ghrelin infusion in healthy volunteers (n = 30), we tested whether ghrelin alters sensitivity to reward and punishment in a reward learning task. Parameters were derived from a computational model of participants' task behavior. The reversal learning task with monetary rewards was performed during functional brain imaging to investigate ghrelin effects on brain signals related to reward prediction errors. Compared to placebo, ghrelin decreased punishment sensitivity (t = -2.448, p = 0.021), while reward sensitivity was unaltered (t = 0.8, p = 0.43). We furthermore found increased prediction-error related activity in the dorsal striatum during ghrelin administration (region of interest analysis: t-values ≥ 4.21, p-values ≤ 0.044). Our results support a role for ghrelin in reward processing that extends beyond food-related rewards. Reduced sensitivity to negative outcomes and increased processing of prediction errors may be beneficial for food foraging when hungry but could also relate to increased risk taking and impulsivity in the broader context of addictive behaviors.


Asunto(s)
Núcleo Caudado , Ghrelina , Castigo , Recompensa , Humanos , Masculino , Ghrelina/farmacología , Ghrelina/administración & dosificación , Método Doble Ciego , Adulto , Adulto Joven , Femenino , Núcleo Caudado/efectos de los fármacos , Núcleo Caudado/diagnóstico por imagen , Núcleo Caudado/metabolismo , Imagen por Resonancia Magnética , Aprendizaje Inverso/efectos de los fármacos , Aprendizaje Inverso/fisiología , Retroalimentación Psicológica/efectos de los fármacos , Retroalimentación Psicológica/fisiología
8.
Addiction ; 119(5): 833-843, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38197836

RESUMEN

BACKGROUND AND AIMS: Total abstinence has historically been the goal of treatment for substance use disorders; however, there is a growing recognition of the health benefits associated with reduced use as a harm reduction measure in stimulant use disorders treatment. We aimed to assess the validity of reduced stimulant use as an outcome measure in randomized controlled trials (RCTs) of pharmacological interventions for stimulant use disorder. DESIGN: We conducted a secondary analysis of a pooled dataset of 13 RCTs. SETTING AND PARTICIPANTS: Participants were individuals seeking treatment for cocaine or methamphetamine use disorders (N = 2062) in a wide range of treatment facilities in the United States. MEASUREMENTS: We validated reduced stimulant use against a set of clinical indicators drawn from harmonized measurements, including severity of problems caused by drug use, comorbid depression, global severity of substance use and improvement, severity of drug-seeking behavior, craving and high-risk behaviors, all assessed at the end of the trial, as well as follow-up urine toxicology. A series of mixed effect regression models was conducted to validate reduction in frequency of use against no reduction in use and abstinence. FINDINGS: More participants reduced frequency of primary drug use than achieved abstinence (18.0% vs. 14.2%, respectively). Reduced use was significantly associated with decreases in craving for the primary drug [60.1%, 95% confidence interval (CI) = 54.3%-64.7%], drug seeking behaviors (41.0%, 95% CI = 36.6%-45.7%), depression severity (39.9%, 95% CI = 30.9%-48.3%), as well as multiple measures of global improvement in psychosocial functioning and severity of drug-related problems, albeit less strongly so than abstinence. Moreover, reduced use was associated with sustained clinical benefit at follow-up, as confirmed by negative urine tests (adjusted odds ratio compared with those with no reduction in use: 0.50, 95% CI = 0.35-0.71). CONCLUSION: Reduced frequency of stimulant use appears to be associated with meaningful improvement in various clinical indicators of recovery. Assessment of reduced use, in addition to abstinence, could broaden the scope of outcomes measured in randomized controlled trials of stimulant use disorders and facilitate the development of more diverse treatment approaches.


Asunto(s)
Estimulantes del Sistema Nervioso Central , Cocaína , Metanfetamina , Trastornos Relacionados con Sustancias , Humanos , Estimulantes del Sistema Nervioso Central/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Trastornos Relacionados con Sustancias/terapia
9.
Drug Alcohol Depend Rep ; 10: 100214, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38234369

RESUMEN

Background: Early use of alcohol and cannabis is associated with health and social problems. It is unclear how lifetime use changes for each additional year of age during adolescence, and whether this change varies by sex and race/ethnicity. This study characterized lifetime rates of alcohol and cannabis use by age among 12- to 17-year-old American youth and explored differential patterns by sex and race/ethnicity. Methods: Data were obtained from the 2019 National Survey on Drug Use and Health. Analyses were restricted to 12-17-year-olds who were non-Hispanic White, non-Hispanic Black, or Hispanic/Latino (n = 11,830). We estimated the increase in lifetime use of alcohol and cannabis by age for the full sample and stratified by sex and race/ethnicity. Slopes of the regression lines were compared to assess differential patterns across groups. Results: In these cross-sectional analyses, reported lifetime use increased substantially from age 12 to 17 for alcohol (6.4 % to 53.2 %) and cannabis (1.3 % to 35.9 %). The increase in lifetime alcohol use was slightly, but not significantly, steeper among girls than boys (F1,8 = 3.40, p = 0.09). White and Latino youth showed similar rates of increase in lifetime alcohol use, which was significantly flatter among Black youth (F2,12=21.26, p<0.0001). Latino youth had a slightly, but not significantly, steeper increase in lifetime cannabis use than White and Black youth (F2,12=3.17, p = 0.07). Conclusions: Reports of lifetime alcohol and cannabis use substantially increase from age 12 to 17 and the rates are different according to sex and race/ethnicity, highlighting the need for early and tailored substance use prevention in adolescents.

10.
Liver Transpl ; 30(2): 200-212, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37934047

RESUMEN

Alcohol-associated liver disease (ALD) has emerged as the leading indication for liver transplantation (LT) worldwide, with 40% of LTs in the United States performed for ALD in 2019. The ALD-related health care burden accelerated during the COVID-19 pandemic, especially in young individuals. Alcohol use disorder (AUD), which focuses on the negative effects of alcohol on psychosocial, physical, and mental health, is present in the majority of patients with ALD, with moderate to severe AUD in 75%-80%. During the last decade, early liver transplantation (eLT) has emerged as a lifesaving treatment for selected patients with alcohol-associated hepatitis; these patients may have a higher risk of using alcohol after LT. The risk of alcohol use recurrence may be reduced during the pretransplant or post-transplant period with AUD treatment using behavioral and/or pharmacological therapies and with regular monitoring for alcohol use (self-reported and complemented with biomarkers like phosphatidylethanol). However, AUD treatment in patients with ALD is challenging due to patient, clinician, and system barriers. An integrated model to provide AUD and ALD care by hepatologists and addiction experts in a colocated clinic starting from LT evaluation and selection to monitoring listed candidates and then to following up on recipients of LT should be promoted. However, the integration of addiction and hepatology teams in an LT program in the real world is often present only during evaluation and candidate selection for LT. Data are emerging to show that a multidisciplinary integrated AUD treatment within an LT program reduces recurrent alcohol use after LT. If we want to continue using early liver transplantation for patients with severe alcohol-associated hepatitis, LT programs should focus on building integrated multidisciplinary care teams for the integrated treatment of both AUD and ALD.


Asunto(s)
Alcoholismo , Hepatitis Alcohólica , Hepatopatías Alcohólicas , Trasplante de Hígado , Humanos , Alcoholismo/complicaciones , Alcoholismo/terapia , Pandemias , Trasplante de Hígado/efectos adversos , Hepatopatías Alcohólicas/complicaciones , Hepatopatías Alcohólicas/cirugía , Hepatitis Alcohólica/complicaciones
11.
Alcohol Alcohol ; 59(1)2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-37665023

RESUMEN

AIMS: The microbiome is a critical factor in health throughout human development. The aims of this scoping review are to (i) elucidate the differences between the youth (post-natal day 21-65 for rodents, 2-7 years for non-human primates, and 10-25 years for humans) microbiome with other life stages and (ii) identify youth-specific microbial changes associated with substance use. METHODS: Peer-reviewed studies published up to May 2023 were identified in PubMed and SCOPUS and included gut and oral microbiome studies from rodents, non-human primates, and humans (N = 1733). Twenty-six articles were determined eligible based on inclusion criteria (aim 1: n = 19, aim 2: n = 7). RESULTS: The adolescent and young adult oral and gut microbiomes are distinct compared to other life stages, within both non-human and human models. While there is limited research in this area, the microbiome appears to be vulnerable to substance use exposure earlier in life, including substances commonly initiated and escalated during adolescence and young adulthood (i.e. alcohol, cannabis, and tobacco). CONCLUSIONS: Studies across the lifespan indicate that adolescence and young adulthood are distinct periods of development, where the microbiome is sensitive to exposures, including substance use. There is a need for more studies focused on the adolescent and young adult microbiome and substance use, as well as focused on the oral microbiome during this developmental period. Understanding the gut and oral microbiome during adolescence and young adulthood may provide insight into the pathophysiology of substance use disorders.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Trastornos Relacionados con Sustancias , Humanos , Adolescente , Adulto Joven , Animales , Adulto , Primates
13.
BMJ Ment Health ; 26(1)2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-37993282

RESUMEN

BACKGROUND: The correlates and consequences of stigma surrounding alcohol use are complex. Alcohol use disorder (AUD) is typically accompanied by self-stigma, due to numerous factors, such as shame, guilt and negative stereotypes. Few studies have empirically examined the possible association between self-stigma and alcohol-related outcomes. OBJECTIVE: To investigate the relationship between self-stigma about alcohol dependence and the severity of alcohol consumption and craving. METHODS: In a sample of 64 participants, the majority of whom had a diagnosis of AUD (51), bivariate correlations were first conducted between Self-Stigma and Alcohol Dependence Scale (SSAD-Apply subscale) scores and Alcohol Use Disorders Identification Test (AUDIT) scores, Alcohol Timeline Follow-Back, Obsessive-Compulsive Drinking Scale (OCDS) scores and Penn Alcohol Cravings Scale scores. Based on the results, regression analyses were conducted with SSAD scores as the predictor and AUDIT and OCDS scores as the outcomes. FINDINGS: SSAD scores positively correlated with AUDIT scores, average drinks per drinking day, number of heavy drinking days and OCDS scores (p<0.001, p=0.014, p=0.011 and p<0.001, respectively). SSAD scores were also found to be a significant predictor of AUDIT and OCDS scores (p<0.001 and p<0.001, respectively), even after controlling for demographics. CONCLUSIONS: Higher levels of self-stigma were associated with more severe AUD, greater alcohol consumption, and more obsessive thoughts and compulsive behaviours related to alcohol. CLINICAL IMPLICATIONS: Our results suggest that potential interventions to reduce self-stigma may lead to improved quality of life and treatment outcomes for individuals with AUD.


Asunto(s)
Alcoholismo , Humanos , Alcoholismo/diagnóstico , Ansia , Calidad de Vida , Consumo de Bebidas Alcohólicas/epidemiología , Conducta Compulsiva/diagnóstico
14.
Transl Psychiatry ; 13(1): 318, 2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37833300

RESUMEN

Alcohol use disorder (AUD) affects transcriptomic, epigenetic and proteomic expression in several organs, including the brain. There has not been a comprehensive analysis of altered protein abundance focusing on the multiple brain regions that undergo neuroadaptations occurring in AUD. We performed a quantitative proteomic analysis using a liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis of human postmortem tissue from brain regions that play key roles in the development and maintenance of AUD, the amygdala (AMG), hippocampus (HIPP), hypothalamus (HYP), nucleus accumbens (NAc), prefrontal cortex (PFC) and ventral tegmental area (VTA). Brain tissues were from adult males with AUD (n = 11) and matched controls (n = 16). Across the two groups, there were >6000 proteins quantified with differential protein abundance in AUD compared to controls in each of the six brain regions. The region with the greatest number of differentially expressed proteins was the AMG, followed by the HYP. Pathways associated with differentially expressed proteins between groups (fold change > 1.5 and LIMMA p < 0.01) were analyzed by Ingenuity Pathway Analysis (IPA). In the AMG, adrenergic, opioid, oxytocin, GABA receptor and cytokine pathways were among the most enriched. In the HYP, dopaminergic signaling pathways were the most enriched. Proteins with differential abundance in AUD highlight potential therapeutic targets such as oxytocin, CSNK1D (PF-670462), GABAB receptor and opioid receptors and may lead to the identification of other potential targets. These results improve our understanding of the molecular alterations of AUD across brain regions that are associated with the development and maintenance of AUD. Proteomic data from this study is publicly available at www.lmdomics.org/AUDBrainProteomeAtlas/ .


Asunto(s)
Alcoholismo , Masculino , Adulto , Humanos , Alcoholismo/metabolismo , Oxitocina , Proteómica , Cromatografía Liquida , Espectrometría de Masas en Tándem , Encéfalo/metabolismo , Proteínas
15.
Res Sq ; 2023 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-37886546

RESUMEN

The stomach-derived hormone ghrelin regulates essential physiological functions. The ghrelin receptor (GHSR) has ligand-independent actions, therefore, GHSR gene deletion may be a reasonable approach to investigate the role of this system in feeding behaviors and diet-induced obesity (DIO). Here we investigated the effects of a long-term (12 month) high-fat (HFD) versus regular diet on obesity-related measures in global GHSR-KO and wild type (WT) Wistar male and female rats. Our main findings were that the GHSR gene deletion protects against DIO and decreases food intake during HFD in male but not in female rats. GHSR gene deletion increased thermogenesis and brain glucose uptake in male rats and modified the effects of HFD on brain glucose metabolism in a sex-specific manner, as assessed with small animal positron emission tomography. RNA-sequencing was also used to show that GHSR-KO rats had upregulated expression of genes responsible for fat oxidation in brown adipose tissue. Central administration of a novel GHSR inverse agonist, PF-5190457, attenuated ghrelin-induced food intake, but only in male, not in female mice. HFD-induced binge-like eating was reduced by inverse agonism in both sexes. Our results support GHSR as a promising target for new pharmacotherapies for obesity.

16.
Front Psychiatry ; 14: 1266424, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37810604

RESUMEN

Background: We conducted a review of all studies comparing clinical aspects of alcohol withdrawal syndrome (AWS) between men and women. Methods: Five databases (PubMed, Cochrane, EMBASE, Scopus and Clinical Trials) were searched for clinical studies using the keywords "alcohol withdrawal syndrome" or "delirium tremens" limited to "sex" or "gender" or "sex difference" or "gender difference." The search was conducted on May 19, 2023. Two reviewers selected studies including both male and female patients with AWS, and they compared males and females in type of AWS symptoms, clinical course, complications, and treatment outcome. Results: Thirty-five observational studies were included with a total of 318,730 participants of which 75,346 had AWS. In twenty of the studies, the number of patients presenting with or developing AWS was separated by sex, resulting in a total of 8,159 (12.5%) female patients and a total of 56,928 (87.5%) male patients. Despite inconsistent results, males were more likely than females to develop complicated AWS [delirium tremens (DT) and AW seizures, collective DT in Males vs. females: 1,792 (85.4%) vs. 307 (14.6%), and collective seizures in males vs. females: 294 (78%) vs. 82 (22%)]. The rates of ICU admissions and hospital length of stay did not show sex differences. Although variable across studies, compared to females, males received benzodiazepine treatment at higher frequency and dose. One study reported that the time from first hospitalization for AWS to death was approximately 1.5 years shorter for males and males had higher mortality rate [19.5% (197/1,016)] compared to females [16% (26/163)]. Conclusion: Despite the significant heterogeneity of the studies selected and the lack of a focus on investigating potential sex differences, this review of clinical studies on AWS suggests that men and women exhibit different AWS manifestations. Large-scale studies focusing specifically on investigating sex difference in AWS are needed.

17.
Neurosci Biobehav Rev ; 154: 105427, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37858908

RESUMEN

Preclinical and human studies suggest a role of aldosterone and mineralocorticoid receptor (MR) in addiction. This scoping review aimed to summarize (1) the relationship between alcohol and other substance use disorders (ASUDs) and dysfunctions of the aldosterone and MR, and (2) how pharmacological manipulations of MR may affect ASUD-related outcomes. Our search in four databases (MEDLINE, Embase, Web of Science, and Cochrane Library) indicated that most studies focused on the relationship between aldosterone, MR, and alcohol (n = 30), with the rest focused on opioids (n = 5), nicotine (n = 9), and other addictive substances (n = 9). Despite some inconsistencies, the overall results suggest peripheral and central dysregulations of aldosterone and MR in several species and that these dysregulations depended on the pattern of drug exposure and genetic factors. We conclude that MR antagonism may be a promising target in ASUD, yet future studies are warranted.


Asunto(s)
Aldosterona , Receptores de Mineralocorticoides , Humanos , Aldosterona/farmacología , Aldosterona/fisiología , Receptores de Mineralocorticoides/genética , Espironolactona/farmacología , Antagonistas de Receptores de Mineralocorticoides/farmacología
18.
Hepatology ; 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37862466

RESUMEN

Alcohol use disorder remains a significant public health concern, affecting around 5% of adults worldwide. Novel pathways of damage have been described during the last years, providing insight into the mechanism of injury due to alcohol misuse beyond the direct effect of ethanol byproducts on the liver parenchyma and neurobehavioral mechanisms. Thus, the gut-liver-brain axis and immune system involvement could be therapeutic targets for alcohol use disorder. In particular, changes in gut microbiota composition and function, and bile acid homeostasis, have been shown with alcohol consumption and cessation. Alcohol can also directly disrupt intestinal and blood-brain barriers. Activation of the immune system can be triggered by intestinal barrier dysfunction and translocation of bacteria, pathogen-associated molecular patterns (such as lipopolysaccharide), cytokines, and damage-associated molecular patterns. These factors, in turn, promote liver and brain inflammation and the progression of liver fibrosis. Other involved mechanisms include oxidative stress, apoptosis, autophagy, and the release of extracellular vesicles and miRNA from hepatocytes. Potential therapeutic targets include gut microbiota (probiotics and fecal microbiota transplantation), neuroinflammatory pathways, as well as neuroendocrine pathways, for example, the ghrelin system (ghrelin receptor blockade), incretin mimetics (glucagon-like peptide-1 analogs), and the mineralocorticoid receptor system (spironolactone). In addition, support with psychological and behavioral treatments is essential to address the multiple dimensions of alcohol use disorder. In the future, a personalized approach considering these novel targets can contribute to significantly decreasing the alcohol-associated burden of disease.

20.
Addict Biol ; 28(7): e13288, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37369125

RESUMEN

Preclinical and clinical work suggests that mifepristone may be a viable treatment for alcohol use disorder (AUD). This was a Phase 1/2, outpatient, cross-over, randomized, double-blind, placebo-controlled trial with non-treatment-seeking individuals with AUD (N = 32). We assessed safety, alcohol craving and consumption, after 1-week mifepristone 600 mg/day administration, in a human laboratory study comprised of a single oral yohimbine administration (32.4 mg), a cue-reactivity procedure and alcohol self-administration. Safety was monitored by adverse events and hemodynamic parameters, alcohol craving by alcohol craving questionnaire and cue-induced saliva output. During the alcohol self-administration, we assessed alcohol pharmacokinetics, subjective effects and consumption. Outcomes were assessed using Generalized Estimating Equations and mediation analysis. Mild-moderate adverse events were reported in both conditions. There was no statistically significant difference between mifepristone and placebo in alcohol pharmacokinetics and subjective effects. Furthermore, blood pressure increased only in the placebo condition after the stress-induced laboratory procedures. Mifepristone, compared to placebo, significantly reduced alcohol craving and increased cortisol levels. Mifepristone-induced cortisol increase was not a mediator of alcohol craving. Mifepristone, compared to placebo, did not reduce alcohol consumption in the laboratory or in a naturalistic setting. This study successfully translated a developed preclinical procedure to a human laboratory study, confirming the safety of mifepristone in people with AUD and providing evidence to its role in reducing alcohol craving under stress procedures. The lack of effects on alcohol drinking may be related to the selection of non-treatment seekers and suggests future treatment-oriented trials should investigate mifepristone in people with AUD.


Asunto(s)
Alcoholismo , Ansia , Humanos , Mifepristona/farmacología , Mifepristona/uso terapéutico , Hidrocortisona/farmacología , Alcoholismo/tratamiento farmacológico , Consumo de Bebidas Alcohólicas , Etanol/farmacología , Método Doble Ciego
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA