Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Am J Drug Alcohol Abuse ; 48(2): 165-175, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35377277

RESUMEN

Background: Hyperpolarization-Activated Cyclic Nucleotide-Gated (HCN) ionic channels are known to play a key role in the control of neuron excitability and have been proposed as a molecular target of ethanol. Previous studies in rats have shown that gene-induced overexpression of the HCN2 channel in the ventral tegmental area (VTA) increases the rewarding effects of ethanol and its intake by the animals.Objective: The aim of this work was to study the effects of VTA HCN2 gene knockdown in the voluntary ethanol consumption of alcohol-preferring UChB rats.Methods: Two lentiviral vectors were generated; LV-siRNA-HCN2, coding for a siRNA that elicited >95% reduction of HCN2 protein levels in vitro, and a control vector coding for a scrambled siRNA sequence. Female UChB naïve rats (n = 14) were microinjected into the VTA with LV-siRNA-HCN2 or the scrambled control vector (n = 11). Four days after, animals were given a daily free access to 10% ethanol and water for 10 days.Results: Rats treated with the LV-siRNA-HCN2 vector showed a ~ 70% reduction (p < .001) in their ethanol preference and ethanol intake compared to control animals. No changes were observed in the total fluid intake of both groups. HCN2 levels in the VTA were measured by Western blot showing a reduction of 40% (p < .05) in the rats injected with LV-siRNA-HCN2, compared to control animals.Conclusion: These results show that knockdown of HCN2 ionic channels in the VTA of UChB rats markedly reduces their voluntary ethanol intake, supporting the idea that HCN2 channels may constitute a therapeutic target for alcohol use disorders.


Asunto(s)
Alcoholismo , Área Tegmental Ventral , Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/metabolismo , Alcoholismo/genética , Animales , Etanol/farmacología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Canales de Potasio/genética , Canales de Potasio/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Área Tegmental Ventral/metabolismo
2.
Adicciones ; 32(3): 208-215, 2020 Jul 01.
Artículo en Inglés, Español | MEDLINE | ID: mdl-31018006

RESUMEN

After ethanol consumption, disulfiram increases blood-acetaldehyde levels, generating an aversive reaction that deters alcohol drinking. Given the major secondary effects of disulfiram, finding other effective drugs to reduce alcohol consumption in individuals with alcohol-use-disorder is highly desirable. It has been reported that administering fenofibrate to high-drinking rats increases hepatic catalase levels and blood acetaldehyde after administering ethanol and a 60-70% inhibition of voluntary alcohol intake. This work evaluated whether fenofibrate has an additional effect on the activity of other ethanol-metabolizing enzymes, which could contribute to the high acetaldehyde levels generated upon administering ethanol. Male high-drinker rats were allowed to voluntary drink 10% ethanol or water for 2 months. Subsequently, fenofibrate (100 mg/kg/day) or vehicle was administered orally for 14 days. Then, alcohol dehydrogenase (ADH1) and aldehyde dehydrogenase (ALDH2) protein levels and enzymatic activities in the livers were quantified. Fenofibrate treatment produced a marked increase in ADH1 protein levels (396% ± 18%, p < 0.001) and enzymatic activity (425% ± 25%, p < 0.001). Fenofibrate did not result in differences in ALDH2 activity or in ALDH2 protein levels. The studies show that treatment with fenofibrate not only increased the activity of catalase in the liver of alcohol-drinking rats, as reported earlier, but also increased the levels and enzymatic activity of ADH1, while ALDH2 remained unchanged. The increases in ADH1 contribute to explaining the remarkable effect of fenofibrate in raising blood levels of acetaldehyde in ethanol-consuming animals, in which a marked reduction of alcohol intake is recorded.


Tras consumir etanol, el disulfiram incrementa los niveles de acetaldehído en sangre y genera una reacción aversiva que desalienta el consumo de alcohol. Dados los importantes efectos secundarios del disulfiram, es altamente deseable hallar otros fármacos efectivos para tratar el trastorno por uso de alcohol. Se ha reportado que administrar fenofibrato a ratas altamente bebedoras de alcohol aumenta los niveles de catalasa hepática y acetaldehído en sangre después de la administración de etanol, y disminuye el consumo voluntario de alcohol (60-70%). Este trabajo evalúa si el fenofibrato tiene un efecto adicional sobre la actividad de otras enzimas en el metabolismo del etanol que podría contribuir a generar altos niveles de acetaldehído. Se permitió a ratas macho altamente bebedoras beber voluntariamente etanol 10% durante 2 meses. Después, se les administró oralmente fenofibrato (100 mg/kg/día) o solo vehículo durante 14 días. Tras eso, se midieron los niveles hepáticos y actividades enzimáticas de alcohol deshidrogenasa (ADH1) y de aldehído deshidrogenasa (ALDH2). El fenofibrato produjo un marcado aumento en los niveles proteicos de ADH1 (396% ± 18%, p < 0,001) y de actividad enzimática (425% ± 25%, p < 0,001) sin alterar los niveles protéicos ni la actividad de ALDH2. Los resultados muestran que el tratamiento con fenofibrato no solo aumenta la actividad de catalasa en el hígado de ratas bebedoras de alcohol, sino que también incrementa los niveles y la actividad de ADH1, sin alterar ALDH2. Esto contribuye a explicar el notable efecto del fenofibrato en aumentar los niveles de acetaldehído en sangre en animales bebedores de alcohol, en los que se registra una marcada reducción en la ingesta de etanol.


Asunto(s)
Alcohol Deshidrogenasa/metabolismo , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Fenofibrato/farmacología , Hipolipemiantes/farmacología , PPAR alfa/agonistas , Acetaldehído/sangre , Aldehído Deshidrogenasa/metabolismo , Animales , Etanol/administración & dosificación , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratas , Ratas Wistar
3.
Gene Ther ; 26(10-11): 407-417, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30820030

RESUMEN

Studies reviewed show that lentiviral gene therapy directed either at inhibiting the synthesis of brain acetaldehyde generated from ethanol or at degrading brain acetaldehyde fully prevent ethanol intake by rats bred for their high alcohol preference. However, after animals have chronically consumed alcohol, the above gene therapy did not inhibit alcohol intake, indicating that in the chronic ethanol intake condition brain acetaldehyde is no longer the compound that generates the continued alcohol reinforcement. Oxidative stress and neuroinflammation generated by chronic ethanol intake are strongly associated with the perpetuation of alcohol consumption and alcohol relapse "binge drinking". Mesenchymal stem cells, referred to as guardians of inflammation, release anti-inflammatory cytokines and antioxidant products. The intravenous delivery of human mesenchymal stem cells or the intranasal administration of mesenchymal stem cell-generated exosomes reverses both (i) alcohol-induced neuro-inflammation and (ii) oxidative stress, and greatly (iii) inhibits (80-90%) chronic alcohol intake and relapse binge-drinking. The therapeutic effect of mesenchymal stem cells is mediated by increased levels of the brain GLT-1 glutamate transporter, indicating that glutamate signaling is pivotal for alcohol relapse. Human mesenchymal stem cells and the products released by these cells may have translational value in the treatment of alcohol-use disorders.


Asunto(s)
Alcoholismo/terapia , Consumo Excesivo de Bebidas Alcohólicas/terapia , Terapia Genética/métodos , Investigación Biomédica Traslacional/métodos , Animales , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos
4.
Alcohol Clin Exp Res ; 40(5): 1044-51, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27062046

RESUMEN

BACKGROUND: A number of studies have shown that acetaldehyde synthesized in the brain is necessary to induce ethanol (EtOH) reinforcement in naïve animals (acquisition phase). However, after chronic intake is achieved (maintenance phase), EtOH intake becomes independent of acetaldehyde generation or its levels. Glutamate has been reported to be associated with the maintenance of chronic EtOH intake. The levels of brain extracellular glutamate are modulated by 2 glial processes: glutamate reabsorption via an Na(+) -glutamate transporter (GLT1) and a cystine-glutamate exchanger. Chronic EtOH intake lowers GLT1 levels and increases extracellular glutamate. The administration of N-acetyl cysteine (NAC), a precursor of cystine, has been shown to reduce the relapse of several drugs of abuse, while NAC has not been tested on chronic EtOH intake or on EtOH's influence on the motivation for another drug. These were investigated in the present study. METHODS: (i) Rats bred for their high EtOH intake were allowed access to 10% EtOH and water up to 87 days. NAC was administered (30 and 60 mg/kg daily, intraperitoneally) for 14 consecutive days, either during the acquisition phase or the maintenance phase of EtOH drinking. (ii) In additional experiments, rats were allowed EtOH (10%) and water access for 61 days, after which EtOH was replaced by saccharin (0.3%) to determine both if chronic EtOH consumption influences saccharin intake and whether NAC modifies the post chronic EtOH saccharin intake. RESULTS: NAC did not influence the acquisition ("first hit") of chronic EtOH intake, but greatly inhibited (60 to 70%; p < 0.0001) EtOH intake when NAC was administered to animals that were consuming EtOH chronically. NAC did not influence saccharin intake in naïve animals. In animals that had consumed EtOH chronically and were thereafter offered a saccharin solution (0.3%), saccharin intake increased over 100% versus that of EtOH-untreated animals, an effect that was fully suppressed by NAC. CONCLUSIONS: N-acetyl cysteine, a drug approved for use in humans, markedly reduces chronic EtOH intake and abolishes the increased intake of saccharin stimulated by chronic EtOH drinking.


Asunto(s)
Acetilcisteína/uso terapéutico , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Motivación/efectos de los fármacos , Sacarina/administración & dosificación , Animales , Masculino , Ratas , Autoadministración , Factores de Tiempo
5.
Addict Biol ; 21(6): 1063-1071, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26032572

RESUMEN

Ethanol is oxidized in the brain to acetaldehyde, which can condense with dopamine to generate (R/S)-salsolinol [(RS)-SAL]. Racemic salsolinol [(RS)-SAL] is self-infused by rats into the posterior ventral tegmental area (VTA) at significantly lower concentrations than those of acetaldehyde, suggesting that (RS)-SAL is a most active product of ethanol metabolism. Early studies showed that repeated intraperitoneal or intra-VTA administration of (RS)-SAL (10 mg/kg) induced conditioned place preference, led to locomotor sensitization and increased voluntary ethanol consumption. In the present study, we separated the (R)- and (S)-enantiomers from a commercial (RS)-SAL using a high-performance liquid chromatography with electrochemical detection system fitted with a ß-cyclodextrin-modified column. We injected (R)-SAL or (S)-SAL (30 pmol/1.0 µl) into the VTA of naïve UChB rats bred as alcohol drinkers to study whether one or both SAL enantiomers are responsible for the motivated behavioral effects, sensitization and increase in voluntary ethanol intake. The present results show that repeated administration of (R)-SAL leads to (1) conditioned place preference; (2) locomotor sensitization; and (3) marked increases in binge-like ethanol intake. Conversely, (S)-SAL did not influence any of these parameters. Overall, data indicate that (R)-SAL stereospecifically induces motivational effects, behavioral sensitization and increases ethanol intake.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Isoquinolinas/farmacología , Análisis de Varianza , Animales , Condicionamiento Psicológico/efectos de los fármacos , Etanol/administración & dosificación , Etanol/metabolismo , Femenino , Fenómenos de Retorno al Lugar Habitual/efectos de los fármacos , Locomoción/efectos de los fármacos , Motivación/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Ratas Wistar
6.
Alcohol Clin Exp Res ; 39(5): 776-86, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25828063

RESUMEN

This review analyzes literature that describes the behavioral effects of 2 metabolites of ethanol (EtOH): acetaldehyde and salsolinol (a condensation product of acetaldehyde and dopamine) generated in the brain. These metabolites are self-administered into specific brain areas by animals, showing strong reinforcing effects. A wealth of evidence shows that EtOH, a drug consumed to attain millimolar concentrations, generates brain metabolites that are reinforcing at micromolar and nanomolar concentrations. Salsolinol administration leads to marked increases in voluntary EtOH intake, an effect inhibited by mu-opioid receptor blockers. In animals that have ingested EtOH chronically, the maintenance of alcohol intake is no longer influenced by EtOH metabolites, as intake is taken over by other brain systems. However, after EtOH withdrawal brain acetaldehyde has a major role in promoting binge-like drinking in the condition known as the "alcohol deprivation effect"; a condition seen in animals that have ingested alcohol chronically, are deprived of EtOH for extended periods, and are allowed EtOH re-access. The review also analyzes the behavioral effects of acetate, a metabolite that enters the brain and is responsible for motor incoordination at low doses of EtOH. Also discussed are the paradoxical effects of systemic acetaldehyde. Overall, evidence strongly suggests that brain-generated EtOH metabolites play a major role in the early ("first-hit") development of alcohol reinforcement and in the generation of relapse-like drinking.


Asunto(s)
Acetaldehído/metabolismo , Acetaldehído/farmacología , Encéfalo/metabolismo , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Etanol/metabolismo , Etanol/farmacología , Isoquinolinas/metabolismo , Refuerzo en Psicología , Acetaldehído/administración & dosificación , Acetatos/farmacología , Animales , Encéfalo/efectos de los fármacos , Isoquinolinas/administración & dosificación , Isoquinolinas/farmacología , Autoadministración
7.
Addict Biol ; 20(2): 336-44, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24571199

RESUMEN

Previous studies suggest that acetaldehyde generated from ethanol in the brain is reinforcing. The present studies tested the feasibility of achieving a long-term reduction of chronic and post-deprivation binge ethanol drinking by a single administration into the brain ventral tegmental area (VTA) of a lentiviral vector that codes for aldehyde dehydrogenase-2 (ALDH2), which degrades acetaldehyde. The ALDH2 gene coding vector or a control lentiviral vector were microinjected into the VTA of rats bred for their alcohol preference. In the chronic alcohol administration model, naïve animals administered the control vector and subsequently offered 10% ethanol and water ingested 8-9 g ethanol/kg body weight/day. The single administration of the ALDH2-coding vector prior to allowing ethanol availability reduced ethanol drinking by 85-90% (P < 0.001) for the 45 days tested. In the post-deprivation binge-drinking model, animals that had previously consumed ethanol chronically for 81 days were administered the lentiviral vector and were thereafter deprived of ethanol for three 7-day periods, each interrupted by a single 60-minute ethanol re-access after the last day of each deprivation period. Upon ethanol re-access, control vector-treated animals consumed intoxicating 'binge' amounts of ethanol, reaching intakes of 2.7 g ethanol/kg body weight in 60 minutes. The administration of the ALDH2-coding vector reduced re-access binge drinking by 75-80% (P < 0.001). This study shows that endowing the ventral tegmental with an increased ability to degrade acetaldehyde greatly reduces chronic alcohol consumption and post-deprivation binge drinking for prolonged periods and supports the hypothesis that brain-generated acetaldehyde promotes alcohol drinking.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Aldehído Deshidrogenasa/genética , Consumo Excesivo de Bebidas Alcohólicas/genética , Proteínas Mitocondriales/genética , Área Tegmental Ventral/metabolismo , Acetaldehído/metabolismo , Consumo de Bebidas Alcohólicas/metabolismo , Alcoholismo/genética , Alcoholismo/metabolismo , Aldehído Deshidrogenasa/metabolismo , Aldehído Deshidrogenasa Mitocondrial , Animales , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Comportamiento de Búsqueda de Drogas , Vectores Genéticos , Lentivirus , Proteínas Mitocondriales/metabolismo , Ratas , Refuerzo en Psicología
8.
Alcohol Clin Exp Res ; 38(4): 911-20, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24460767

RESUMEN

BACKGROUND: A number of studies have shown that ethanol (EtOH) activates dopamine neurocircuitries and is self-administered into the ventral tegmental area (VTA) of the rat brain. In vitro and in silico studies have showed that hyperpolarization-activated cyclic nucleotide-gated (HCN) ionic channels on VTA dopamine neurons may constitute a molecular target of EtOH; however, there is no in vivo evidence supporting this assumption. METHODS: Wistar-derived University of Chile Drinking (UChB) rats were microinjected into the VTA with a lentiviral vector coding for rat HCN-2 ionic channel or a control vector. Four days after vector administration, daily voluntary EtOH intake was assessed for 30 days under a free-access paradigm to 5% EtOH and water. After EtOH consumption studies, the effect of HCN-2 overexpression was also assessed on EtOH-induced conditioned place preference (CPP); EtOH-induced locomotion, and EtOH-induced dopamine release in the nucleus accumbens (NAcc). RESULTS: Rats microinjected with the HCN-2 coding vector into the VTA showed (i) a ~2-fold increase in their voluntary EtOH intake compared to control animals, (ii) lentiviral-HCN-2-treated animals also showed an increased CPP to EtOH (~3-fold), (iii) a significant higher locomotor activity (~2-fold), and (iv) increased dopamine release in NAcc upon systemic administration of EtOH (~2-fold). CONCLUSIONS: Overexpression of HCN-2 ionic channel in the VTA of rats results in an increase in voluntary EtOH intake, EtOH-induced CPP, locomotor activity, and dopamine release in NAcc, suggesting that HCN levels in the VTA are relevant for the rewarding properties of EtOH.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Etanol/administración & dosificación , Regulación de la Expresión Génica , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Canales de Potasio/biosíntesis , Recompensa , Área Tegmental Ventral/metabolismo , Animales , Femenino , Células HEK293 , Humanos , Ratas , Ratas Wistar , Autoadministración , Área Tegmental Ventral/efectos de los fármacos
9.
Artículo en Inglés | MEDLINE | ID: mdl-38565388

RESUMEN

While there is extensive research on alcohol dependence, the factors that make an individual vulnerable to developing alcoholism haven't been explored much. In this study, we aim to investigate how neonatal exposure to sex hormones affects alcohol intake and the regulation of the mesolimbic pathway in adulthood. The study aimed to investigate the impact of neonatal exposure to a single dose of testosterone propionate (TP) or estradiol valerate (EV) on ethanol consumption in adult rats. The rats were subjected to a two-bottle free-choice paradigm, and the content of dopamine (DA) and 3,4-dihydroxyphenylacetic acid (DOPAC) in the nucleus accumbens (NAcc) was measured using HPLC-ED. The expression of critical DA-related proteins in the mesolimbic pathway was evaluated through RT-qPCR and western blot analysis. Supraphysiological neonatal exposure to EV or TP resulted in increased ethanol intake over four weeks in adulthood. In addition, the DA and DOPAC content was reduced and increased in the NAcc of EV and TP-treated rats, and ß-endorphin content in the hypothalamus decreased in EV-treated rats. The VTA µ receptor and DA type 2 form short receptor (D2S) expression were significantly reduced in EV and TP male rats. Finally, in an extended 6-week protocol, the increase in ethanol consumption induced by EV was mitigated during the initial two hours post-naloxone injection. Neonatal exposure to sex hormones is a detrimental stimulus for the brain, which can facilitate the development of addictive behaviors, including alcohol use disorder.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético , Consumo de Bebidas Alcohólicas , Animales Recién Nacidos , Dopamina , Estradiol , Núcleo Accumbens , Propionato de Testosterona , Animales , Masculino , Consumo de Bebidas Alcohólicas/metabolismo , Ratas , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efectos de los fármacos , Estradiol/farmacología , Propionato de Testosterona/farmacología , Propionato de Testosterona/administración & dosificación , Dopamina/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Femenino , Etanol/farmacología , Etanol/administración & dosificación , Hormonas Esteroides Gonadales/metabolismo , Ratas Sprague-Dawley
10.
Alcohol Clin Exp Res ; 37(8): 1278-85, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23527889

RESUMEN

BACKGROUND: Animals that have chronically consumed alcohol and are subsequently deprived of it markedly increase their intake above basal levels when access to alcohol is reinstated. Such an effect, termed the alcohol deprivation effect (ADE), has been proposed to reflect (i) an obsessive-compulsive behavior, (ii) craving, or (iii) an increased reinforcing value of ethanol (EtOH). It has been reported that acetaldehyde, a highly reinforcing metabolite of EtOH, is generated in the brain by the action of catalase. Recent studies show that the administration of an anticatalase (shRNA)-encoding lentiviral vector into the brain ventral tegmental area (VTA) of naïve rats virtually abolishes (85 to 95%) their EtOH intake. It is hypothesized that the antireinforcing effect of the anticatalase vector will also inhibit the ADE. METHODS: Two-month-old Wistar-derived UChB alcohol drinker rats were offered free access to water and 10 and 20% EtOH for 67 days. Thereafter, the animals were deprived of EtOH for 15 days and were subsequently offered access to the EtOH solutions. At the start of the deprivation period, animals were microinjected a single dose of an anticatalase (or control) vector into the VTA. EtOH intake was measured on the first hour of EtOH re-exposure as well as on a 24-hour basis for 7 days. RESULTS: A marked ADE was observed when EtOH intake was measured on the first hour or 24 hours following EtOH re-exposure, compared to the corresponding controls. The administration of the anticatalase vector reduced ADE by 60 to 80% (p < 0.001) on the first hour and by 63 to 80% (p < 0.001) on the initial 24 hours of EtOH re-exposure (first and second ADE, respectively) without changing the total fluid intake, indicating a specific effect on EtOH drinking. CONCLUSIONS: Ethanol intake associated with ADE--a binge-like drinking behavior--is markedly inhibited by the administration of an anticatalase vector into the VTA, which blocks the conversion of EtOH into acetaldehyde, strongly suggesting that the marked increased EtOH intake that follows an alcohol deprivation period is mediated by acetaldehyde and its reinforcing metabolite.


Asunto(s)
Acetaldehído/metabolismo , Consumo de Bebidas Alcohólicas/metabolismo , Trastornos Relacionados con Alcohol/enzimología , Catalasa/antagonistas & inhibidores , Área Tegmental Ventral/enzimología , Consumo de Bebidas Alcohólicas/prevención & control , Trastornos Relacionados con Alcohol/prevención & control , Animales , Catalasa/metabolismo , Femenino , Terapia Genética , Ratas , Ratas Wistar
11.
Drug Alcohol Depend ; 250: 110900, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37515828

RESUMEN

RATIONALE: Neuronal nicotinic acetylcholine receptors (nAChRs) are implicated in the reinforcing effects of nicotine and ethanol. Previous studies have shown that cytisine and its 5-bromo derivative are partial agonists at the α4ß2 nAChRs and that the parent molecule cytisine is effective in reducing both nicotine- and ethanol-self-administration in rats. However, whether 5-bromocytisine affects nicotine or ethanol self-administration was unknown. OBJECTIVES: The present study compared the effects of 5-bromocytisine and cytisine on nicotine self-administration and further assessed the effect of daily drug injection on voluntary ethanol consumption in alcohol-preferring female rats. Animals were administered a 1.5mg/kg i.p. dose of 5-bromocytisine or cytisine every day for 15-16 days. RESULTS: The initial efficacy of 5-bromocytisine and cytisine in reducing nicotine intake was similar (-80%) while for voluntary ethanol intake 5-bromocytisine was a superior inhibitor over cytisine (-78% and -40% respectively). The efficacy of cytisine began to diminish after 10 days of daily administration, which was attributed to tolerance development to its inhibitory effects both on nicotine and ethanol self-administration. Tolerance did not develop for 5-bromocytisine. CONCLUSION: 5-Bromocytisine, a weaker α4ß2 nAChR partial agonist than cytisine, also produces a sustained inhibition of both nicotine and ethanol self-administration, and unlike cytisine, it does not develop tolerance.


Asunto(s)
Alcaloides , Receptores Nicotínicos , Ratas , Femenino , Animales , Nicotina/farmacología , Etanol , Alcaloides/farmacología , Agonistas Nicotínicos/farmacología , Hidrocarburos Aromáticos con Puentes/farmacología
12.
Neuropsychopharmacology ; 48(9): 1367-1376, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36175550

RESUMEN

The nucleus accumbens (nAc) is a critical region in the brain reward system since it integrates abundant synaptic inputs contributing to the control of neuronal excitability in the circuit. The presence of inhibitory α1 glycine receptor (GlyRs) subunits, sensitive to ethanol, has been recently reported in accumbal neurons suggesting that they are protective against excessive binge consumption. In the present study, we used viral vectors (AAV) to overexpress mutant and WT α1 subunits in accumbal neurons in D1 Cre and α1 KI mice. Injection of a Cre-inducible AAV carrying an ethanol insensitive α1 subunit in D1 Cre neurons was unable to affect sensitivity to ethanol in GlyRs or affect ethanol drinking. On the other hand, using an AAV that transduced WT α1 GlyRs in GABAergic neurons in the nAc of high-ethanol consuming mice caused a reduction in ethanol intake as reflected by lowered drinking in the dark and reduced blood ethanol concentration. As expected, the AAV increased the glycine current density by 5-fold without changing the expression of GABAA receptors. Examination of the ethanol sensitivity in isolated accumbal neurons indicated that the GlyRs phenotype changed from an ethanol resistant to an ethanol sensitive type. These results support the conclusion that increased inhibition in the nAc can control excessive ethanol consumption and that selective targeting of GlyRs by pharmacotherapy might provide a mechanistic procedure to reduce ethanol binge.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas , Glicina , Animales , Ratones , Consumo Excesivo de Bebidas Alcohólicas/genética , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Etanol/farmacología , Neuronas GABAérgicas/metabolismo , Glicina/farmacología , Glicina/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Glicina/genética , Receptores de Glicina/metabolismo
13.
Life Sci ; 328: 121876, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37348813

RESUMEN

AIMS: Alcohol relapse is a main limitation for the treatment of alcohol use disorders. Previous studies have shown that Alda-1, a pharmacological activator of ALDH2, inhibits both acquisition and chronic ethanol intake in rats; however, its effects on relapse-like ethanol intake are unknown. The aim of this study was to assess the effect of Alda-1 on post-deprivation and reaccess relapse-like ethanol intake in alcohol-preferring UChB rats. We also aimed to assess the possible mechanisms associated with the effects of Alda-1 by measuring the levels of glutamate transporter (GLT-1), oxidative stress and neuroinflammation markers in different regions of the mesocorticolimbic system. MAIN METHODS: In Experiment I, UChB female rats were exposed for 100 days to voluntary ethanol intake followed by 2-weeks of ethanol withdrawal and 1 week of ethanol reaccess. Alda-1 (25 mg/kg, intragastric, i.g) or vehicle was administered daily for 14 days during the withdrawal/re-access period. Experiment II was similar to Experiment I, but after the withdrawal period, ethanol re-access was not allowed, and Alda-1 was administered during the last week of withdrawal. At the end of both experiments, the levels of GLT-1, oxidative stress (GSH, MDA), and neuroinflammation markers (GFAP, Iba-1) were assessed in nucleus accumbens and/or hippocampus. KEY FINDINGS: The results showed that Alda-1 administration markedly blocked (90 %, p < 0.001) relapse-like ethanol intake in UChB rats. Alda-1 increased Iba-1 reactivity (microglial marker) in the NAc of ethanol-deprived rats. Alda-1 administration did not influence the levels of GLT-1, oxidative stress markers (MDA, GSH) or GFAP reactivity in the mesocorticolimbic system. SIGNIFICANCE: These preclinical findings support the use of activators of ALDH2, such as Alda-1, as a potential pharmacological strategy in the treatment of alcohol relapse.


Asunto(s)
Alcoholismo , Etanol , Ratas , Femenino , Animales , Alcoholismo/tratamiento farmacológico , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Enfermedades Neuroinflamatorias , Aldehído Deshidrogenasa Mitocondrial , Enfermedad Crónica , Sistema de Transporte de Aminoácidos X-AG , Recurrencia
14.
ACS Omega ; 8(41): 38566-38576, 2023 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-37867702

RESUMEN

Salsolinol (1-methyl-1,2,3,4-tetrahydroisoquinoline-6,7-diol) is a close structural analogue of dopamine with an asymmetric center at the C1 position, and its presence in vivo, both in humans and rodents, has already been proven. Yet, given the fact that salsolinol colocalizes with dopamine-rich regions and was first detected in the urine of Parkinson's disease patients, its direct role in the process of neurodegeneration has been proposed. Here, we report that R and S enantiomers of salsolinol, which we purified from commercially available racemic mixture by means of high-performance liquid chromatography, exhibited neuroprotective properties (at the concentration of 50 µM) toward the human dopaminergic SH-SY5Y neuroblastoma cell line. Furthermore, within the study, we observed no toxic effect of N-methyl-(R)-salsolinol on SH-SY5Y neuroblastoma cells up to the concentration of 750 µM, either. Additionally, our molecular docking analysis showed that enantiomers of salsolinol should exhibit a distinct ability to interact with dopamine D2 receptors. Thus, we postulate that our results highlight the need to acknowledge salsolinol as an active dopamine metabolite and to further explore the neuroregulatory role of enantiomers of salsolinol.

15.
Neurotoxicology ; 97: 12-24, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37142061

RESUMEN

Lead (Pb), a common environmental contaminant, and ethanol (EtOH), a widely available drug of abuse, are well-known neurotoxicants. In vivo, experimental evidence indicates that Pb exposure affects oxidative EtOH metabolism with a high impact on living organisms. On these bases, we evaluated the consequences of combined Pb and EtOH exposure on aldehyde dehydrogenase 2 (ALDH2) functionality. In vitro exposure to 10 µM Pb, 200 mM EtOH, or their combination for 24 h reduced ALDH2 activity and content in SH-SY5Y human neuroblastoma cells. In this scenario, we observed mitochondrial dysfunction characterized by reduced mass and membrane potential, decreased maximal respiration, and spare capacity. We also evaluated the oxidative balance in these cells finding a significant increase in reactive oxygen species (ROS) production and lipid peroxidation products under all treatments accompanied by an increase in catalase (CAT) activity and content. These data suggest that ALDH2 inhibition induces the activation of converging cytotoxic mechanisms resulting in an interplay between mitochondrial dysfunction and oxidative stress. Notably, NAD+ (1 mM for 24 h) restored ALDH2 activity in all groups, while an ALDH2 enhancer (Alda-1, 20 µM for 24 h) also reversed some of the deleterious effects resulting from impaired ALDH2 function. Overall, these results reveal the crucial role of this enzyme on the Pb and EtOH interaction and the potential of activators such as Alda-1 as therapeutic approaches against several conditions involving aldehydes accumulation.


Asunto(s)
Etanol , Neuroblastoma , Humanos , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Etanol/toxicidad , Plomo/toxicidad , Plomo/metabolismo , Neuroblastoma/metabolismo , Antioxidantes/metabolismo , Oxidación-Reducción , Línea Celular , Mitocondrias/metabolismo , Benzodioxoles
16.
Alcohol Clin Exp Res ; 36(3): 517-22, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21895710

RESUMEN

BACKGROUND: In animal models of continuous alcohol self-administration, in which physical dependence does not constitute the major factor of ethanol intake, 2 factors likely contribute to the perpetuation of alcohol self-administration: (i) the rewarding effects of ethanol and (ii) the contextual conditioning cues that exist along with the process of self-administration. Present studies are aimed at understanding the relative contribution of these factors on the perpetuation of heavy alcohol self-administration, as an indication of relapse. METHODS: Wistar-derived UChB high ethanol drinker rats were allowed access to 10% ethanol and water on a 24-hour basis. In initial studies, an anticatalase shRNA gene-coding lentiviral vector aimed at inhibiting acetaldehyde generation was administered into the ventral tegmental area (VTA) of the animals prior to ethanol access. In subsequent studies, the lentiviral vector was administered to animals, which had consumed ethanol on a 24-hour basis, or a 1-hour basis, after the animals had reached high levels of ethanol intake for 60 to 80 days. In final studies, quinine (0.01%) was added to the ethanol solution to alter the conditioning taste/smell cues of alcohol that animals had chronically ingested. RESULTS: Data indicate that the administration of an anticatalase vector into the VTA of naïve animals blocked reward and alcohol self-administration, while it was, nevertheless, inactive in inhibiting alcohol self-administration in rats that had been conditioned to ingest ethanol for over 2 months. The lack of inhibitory effect of the anticatalase vector on ethanol intake in animals that had chronically self-administered ethanol was fully reversed when the contextual conditioning cues of the alcohol solution were changed. CONCLUSIONS: Data highlight the importance of conditioning factors in relapse and suggest that only abolishing or blunting it, along with long-lasting pharmacological treatment to reduce ethanol reward, may have protracted effects in reducing alcohol self-administration.


Asunto(s)
Consumo de Bebidas Alcohólicas/psicología , Alcoholismo/prevención & control , Alcoholismo/terapia , Recompensa , Acetaldehído/metabolismo , Consumo de Bebidas Alcohólicas/metabolismo , Consumo de Bebidas Alcohólicas/terapia , Alcoholismo/genética , Alcoholismo/metabolismo , Animales , Catalasa/antagonistas & inhibidores , Catalasa/genética , Señales (Psicología) , Modelos Animales de Enfermedad , Etanol/administración & dosificación , Etanol/antagonistas & inhibidores , Etanol/farmacología , Vectores Genéticos/administración & dosificación , Humanos , Lentivirus/genética , Microinyecciones , Quinina/farmacología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/uso terapéutico , Ratas , Ratas Wistar , Prevención Secundaria , Autoadministración , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
17.
Alcohol Alcohol ; 47(2): 102-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22214999

RESUMEN

AIMS: To mimic, in an animal model of alcoholism, the protective phenotype against alcohol consumption observed in humans carrying a fast alcohol dehydrogenase (ADH1B*2) and an inactive aldehyde dehydrogenase (ALDH2*2). METHODS: We developed a multiple expression cassette adenoviral vector (AdV-ADH/asALDH2) encoding both a fast rat ADH and an antisense RNA against rat ALDH2. A control adenoviral vector (AdV-C) containing intronic non-coding DNA was also developed. These adenoviral vectors were administered intravenously to rats bred as high alcohol-drinkers (University of Chile bibulous) that were previously rendered alcohol dependent by a 75-day period of voluntary 10% ethanol intake. RESULTS: Animals administered AdV-ADH/asALDH2 showed a 176% increase in liver ADH activity, whereas liver ALDH2 activity was reduced by 24%, and upon the administration of a dose of ethanol (1 g/kg, i.p.), these showed arterial acetaldehyde levels that were 400% higher than those of animals administered AdV-C. Rats that received the AdV-ADH/asALDH2 vector reduced by 60% their voluntary ethanol intake versus controls. CONCLUSION: This study provides evidence that the simultaneous increase of liver ADH and a reduction of ALDH activity by gene transfer could constitute a potential therapeutic strategy for the treatment of alcoholism.


Asunto(s)
Alcohol Deshidrogenasa/genética , Consumo de Bebidas Alcohólicas/genética , Alcoholismo/terapia , Aldehído Deshidrogenasa/antagonistas & inhibidores , Técnicas de Transferencia de Gen/psicología , Vectores Genéticos/uso terapéutico , Proteínas Mitocondriales/antagonistas & inhibidores , ARN sin Sentido/uso terapéutico , Acetaldehído/sangre , Adenoviridae/genética , Alcohol Deshidrogenasa/metabolismo , Consumo de Bebidas Alcohólicas/sangre , Consumo de Bebidas Alcohólicas/metabolismo , Alcoholismo/sangre , Alcoholismo/genética , Alcoholismo/metabolismo , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa Mitocondrial , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Isoenzimas/genética , Isoenzimas/metabolismo , Hígado/metabolismo , Proteínas Mitocondriales/genética , ARN sin Sentido/genética , Ratas , Ratas Wistar
18.
Biomedicines ; 10(7)2022 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-35884787

RESUMEN

Alcoholism is a worldwide public health problem with high economic cost and which affects health and social behavior. It is estimated that alcoholism kills 3 million people globally, while in Chile it is responsible for around 9 thousand deaths per year. Nicotinic acetylcholine receptors (nAChRs) are ligand-gated ion channels expressed in the central nervous system, and they were suggested to modulate the ethanol mechanism involved in abuse and dependence. Previous work demonstrated a short-term treatment with UFR2709, a nAChRs antagonist, which reduced ethanol intake using a two-bottle free-choice paradigm in University of Chile bibulous (UChB) rats. Here, we present evidence of the UFR2709 efficacy in reducing the acquisition and long-term ethanol consumption. Our results show that UFR2709 (2.5 mg/kg i.p.) reduces the seek behavior and ethanol intake, even when the drug administration was stopped, and induced a reduction in the overall ethanol intake by around 55%. Using naïve UChB bibulous rats, we demonstrate that UFR2709 could delay and reduce the genetically adaptive impulse to seek and drink ethanol and prevent its excessive intake.

19.
Front Immunol ; 13: 1035589, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36713380

RESUMEN

Introduction: Chronic Chagasic cardiomyopathy (CCC), caused by the protozoan Trypanosoma cruzi, is the most severe manifestation of Chagas disease.CCC is characterized by cardiac inflammation and fibrosis caused by a persistent inflammatory response. Following infection, macrophages secrete inflammatory mediators such as IL-1ß, IL-6, and TNF-α to control parasitemia. Although this response contains parasite infection, it causes damage to the heart tissue. Thus, the use of immunomodulators is a rational alternative to CCC. Rho-associated kinase (ROCK) 1 and 2 are RhoA-activated serine/threonine kinases that regulate the actomyosin cytoskeleton. Both ROCKs have been implicated in the polarization of macrophages towards an M1 (pro-inflammatory) phenotype. Statins are FDA-approved lipid-lowering drugs that reduce RhoA signaling by inhibiting geranylgeranyl pyrophosphate (GGPP) synthesis. This work aims to identify the effect of statins on U937 macrophage polarization and cardiac tissue inflammation and its relationship with ROCK activity during T. cruzi infection. Methods: PMA-induced, wild-type, GFP-, CA-ROCK1- and CA-ROCK2-expressing U937 macrophages were incubated with atorvastatin, or the inhibitors Y-27632, JSH-23, TAK-242, or C3 exoenzyme incubated with or without T. cruzi trypomastigotes for 30 min to evaluate the activity of ROCK and the M1 and M2 cytokine expression and secretion profiling. Also, ROCK activity was determined in T. cruzi-infected, BALB/c mice hearts. Results: In this study, we demonstrate for the first time in macrophages that incubation with T. cruzi leads to ROCK activation via the TLR4 pathway, which triggers NF-κB activation. Inhibition of ROCKs by Y-27632 prevents NF-κB activation and the expression and secretion of M1 markers, as does treatment with atorvastatin. Furthermore, we show that the effect of atorvastatin on the NF-kB pathway and cytokine secretion is mediated by ROCK. Finally, statin treatment decreased ROCK activation and expression, and the pro-inflammatory cytokine production, promoting anti-inflammatory cytokine expression in chronic chagasic mice hearts. Conclusion: These results suggest that the statin modulation of the inflammatory response due to ROCK inhibition is a potential pharmacological strategy to prevent cardiac inflammation in CCC.


Asunto(s)
Cardiomiopatías , Enfermedad de Chagas , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Trypanosoma cruzi , Humanos , Animales , Ratones , Trypanosoma cruzi/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Quinasas Asociadas a rho/metabolismo , FN-kappa B/metabolismo , Atorvastatina/farmacología , Células U937 , Macrófagos/metabolismo , Enfermedad de Chagas/genética , Citocinas/metabolismo , Cardiomiopatías/metabolismo , Inflamación/metabolismo
20.
FASEB J ; 24(1): 266-74, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19710201

RESUMEN

Humans who carry a point mutation in the gene coding for alcohol dehydrogenase-1B (ADH1B*2; Arg47His) are markedly protected against alcoholism. Although this mutation results in a 100-fold increase in enzyme activity, it has not been reported to cause higher levels of acetaldehyde, a metabolite of ethanol known to deter alcohol intake. Hence, the mechanism by which this mutation confers protection against alcoholism is unknown. To study this protective effect, the wild-type rat cDNA encoding rADH-47Arg was mutated to encode rADH-47His, mimicking the human mutation. The mutated cDNA was incorporated into an adenoviral vector and administered to genetically selected alcohol-preferring rats. The V(max) of rADH-47His was 6-fold higher (P<0.001) than that of the wild-type rADH-47Arg. Animals transduced with rAdh-47His showed a 90% (P<0.01) increase in liver ADH activity and a 50% reduction (P<0.001) in voluntary ethanol intake. In animals transduced with rAdh-47His, administration of ethanol (1g/kg) produced a short-lived increase of arterial blood acetaldehyde concentration to levels that were 3.5- to 5-fold greater than those in animals transduced with the wild-type rAdh-47Arg vector or with a noncoding vector. This brief increase (burst) in arterial acetaldehyde concentration after ethanol ingestion may constitute the mechanism by which humans carrying the ADH1B*2 allele are protected against alcoholism.


Asunto(s)
Alcohol Deshidrogenasa/genética , Alcoholismo/enzimología , Alcoholismo/prevención & control , Acetaldehído/sangre , Adenoviridae/genética , Alcohol Deshidrogenasa/metabolismo , Alcoholismo/genética , Alelos , Sustitución de Aminoácidos , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Línea Celular , Cartilla de ADN/genética , Modelos Animales de Enfermedad , Etanol/administración & dosificación , Vectores Genéticos , Humanos , Cinética , Mutagénesis Sitio-Dirigida , Mutación Puntual , Polimorfismo Genético , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción Genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA