Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.837
Filtrar
1.
Addict Biol ; 29(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38963015

RESUMEN

The addictive use of nicotine contained in tobacco is associated with stressor-like emotional and cognitive effects such as anxiety and working memory impairment, and the involvement of epigenetic mechanisms such as histone acetylation has recently been reported. Although the precise nature of behavioural plasticity remains unclear, both anxiogenic- and working memory impairment-like effects were observed in the present experimental model of mice treated with repeated subcutaneous nicotine and/or immobilization stress, and these effects were commonly attenuated by the histone deacetylase (HDAC) inhibitors that induce histone acetylation. Such HDAC inhibitor-induced resilience was mimicked by ligands for the endocannabinoid (ECB) system, a neurotransmitter system that is closely associated with nicotine-induced addiction-related behaviours: the anxiogenic-like effects were mitigated by the cannabinoid type 1 (CB1) agonist arachidonylcyclopropylamide (ACPA), whereas the working memory impairment-like effects were mitigated by the CB1 antagonist SR 141716A. Moreover, the effects of the HDAC inhibitors were also mimicked by ligands for the endovanilloid (transient receptor potential vanilloid 1 [TRPV1]) system, a system that shares common characteristics with the ECB system: the anxiogenic-like effects were mitigated by the TRPV1 antagonist capsazepine, whereas the working memory impairment-like effects were mitigated by the TRPV1 agonist olvanil. Notably, the HDAC inhibitor-induced anxiolytic-like effects were attenuated by SR 141716A, which were further counteracted by capsazepine, whereas the working memory improvement-like effects were attenuated by capsazepine, which were further counteracted by SR 141716A. These results suggest the contribution of interrelated control of the ECB/TRPV1 systems and epigenetic processes such as histone acetylation to novel therapeutic approaches.


Asunto(s)
Ansiedad , Endocannabinoides , Epigénesis Genética , Memoria a Corto Plazo , Nicotina , Estrés Psicológico , Canales Catiónicos TRPV , Animales , Canales Catiónicos TRPV/efectos de los fármacos , Nicotina/farmacología , Ratones , Memoria a Corto Plazo/efectos de los fármacos , Endocannabinoides/metabolismo , Masculino , Epigénesis Genética/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Receptor Cannabinoide CB1/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Capsaicina/farmacología , Capsaicina/análogos & derivados , Modelos Animales de Enfermedad , Rimonabant/farmacología , Agonistas Nicotínicos/farmacología , Piperidinas/farmacología
2.
ASN Neuro ; 16(1): 2371160, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39024573

RESUMEN

Promising new pharmacological strategies for the enhancement of cognition target either nicotinic acetylcholine receptors (nAChR) or N-methyl-D-aspartate receptors (NMDAR). There is also an increasing interest in low-dose combination therapies co-targeting the above neurotransmitter systems to reach greater efficacy over the monotreatments and to reduce possible side effects of high-dose monotreatments. In the present study, we assessed modulatory effects of the α7 nAChR-selective agonist PHA-543613 (PHA), a novel α7 nAChR positive allosteric modulator compound (CompoundX) and the NMDAR antagonist memantine on the in vivo firing activity of CA1 pyramidal neurons in the rat hippocampus. Three different test conditions were applied: spontaneous firing activity, NMDA-evoked firing activity and ACh-evoked firing activity. Results showed that high but not low doses of memantine decreased NMDA-evoked firing activity, and low doses increased the spontaneous and ACh-evoked firing activity. Systemically applied PHA robustly potentiated ACh-evoked firing activity with having no effect on NMDA-evoked activity. In addition, CompoundX increased both NMDA- and ACh-evoked firing activity, having no effects on spontaneous firing of the neurons. A combination of low doses of memantine and PHA increased firing activity in all test conditions and similar effects were observed with memantine and CompoundX but without spontaneous firing activity increasing effects. Our present results demonstrate that α7 nAChR agents beneficially interact with Alzheimer's disease medication memantine. Moreover, positive allosteric modulators potentiate memantine effects on the right time and the right place without affecting spontaneous firing activity. All these data confirm previous behavioral evidence for the viability of combination therapies for cognitive enhancement.


Asunto(s)
Hipocampo , Memantina , Receptor Nicotínico de Acetilcolina alfa 7 , Animales , Memantina/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Receptor Nicotínico de Acetilcolina alfa 7/antagonistas & inhibidores , Hipocampo/efectos de los fármacos , Masculino , Ratas , Neuronas/efectos de los fármacos , Neuronas/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Relación Dosis-Respuesta a Droga , Cognición/efectos de los fármacos , Cognición/fisiología , Antagonistas de Aminoácidos Excitadores/farmacología , Nootrópicos/farmacología , Ratas Wistar , Ligandos , Agonistas Nicotínicos/farmacología
3.
Antiviral Res ; 228: 105934, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38880195

RESUMEN

Herpes simplex virus type 1 (HSV-1), a neurotropic DNA virus, establishes latency in neural tissues, with reactivation causing severe consequences like encephalitis. Emerging evidence links HSV-1 infection to chronic neuroinflammation and neurodegenerative diseases. Microglia, the central nervous system's (CNS) immune sentinels, express diverse receptors, including α7 nicotinic acetylcholine receptors (α7 nAChRs), critical for immune regulation. Recent studies suggest α7 nAChR activation protects against viral infections. Here, we show that α7 nAChR agonists, choline and PNU-282987, significantly inhibit HSV-1 replication in microglial BV2 cells. Notably, this inhibition is independent of the traditional ionotropic nAChR signaling pathway. mRNA profiling revealed that choline stimulates the expression of antiviral factors, IL-1ß and Nos2, and down-regulates the apoptosis genes and type A Lamins in BV2 cells. These findings suggest a novel mechanism by which microglial α7 nAChRs restrict viral infections by regulating innate immune responses.


Asunto(s)
Colina , Herpesvirus Humano 1 , Microglía , Replicación Viral , Receptor Nicotínico de Acetilcolina alfa 7 , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/genética , Microglía/virología , Microglía/efectos de los fármacos , Microglía/metabolismo , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 1/efectos de los fármacos , Animales , Línea Celular , Ratones , Replicación Viral/efectos de los fármacos , Colina/farmacología , Colina/metabolismo , Compuestos Bicíclicos con Puentes/farmacología , Benzamidas/farmacología , Inmunidad Innata , Herpes Simple/virología , Herpes Simple/metabolismo , Interleucina-1beta/metabolismo , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Antivirales/farmacología , Agonistas Nicotínicos/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética
4.
Hepatol Commun ; 8(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38836815

RESUMEN

BACKGROUND: Smoking is a risk factor for liver cirrhosis; however, the underlying mechanisms remain largely unexplored. The α7 nicotinic acetylcholine receptor (α7nAChR) has recently been detected in nonimmune cells possessing immunoregulatory functions. We aimed to verify whether nicotine promotes liver fibrosis via α7nAChR. METHODS: We used osmotic pumps to administer nicotine and carbon tetrachloride to induce liver fibrosis in wild-type and α7nAChR-deficient mice. The severity of fibrosis was evaluated using Masson trichrome staining, hydroxyproline assays, and real-time PCR for profibrotic genes. Furthermore, we evaluated the cell proliferative capacity and COL1A1 mRNA expression in human HSCs line LX-2 and primary rat HSCs treated with nicotine and an α7nAChR antagonist, methyllycaconitine citrate. RESULTS: Nicotine exacerbated carbon tetrachloride-induced liver fibrosis in mice (+42.4% in hydroxyproline assay). This effect of nicotine was abolished in α7nAChR-deficient mice, indicating nicotine promotes liver fibrosis via α7nAChR. To confirm the direct involvement of α7nAChRs in liver fibrosis, we investigated the effects of genetic suppression of α7nAChR expression on carbon tetrachloride-induced liver fibrosis without nicotine treatment. Profibrotic gene expression at 1.5 weeks was significantly suppressed in α7nAChR-deficient mice (-83.8% in Acta2, -80.6% in Col1a1, -66.8% in Tgfb1), and collagen content was decreased at 4 weeks (-22.3% in hydroxyproline assay). The in vitro analysis showed α7nAChR expression in activated but not in quiescent HSCs. Treatment of LX-2 cells with nicotine increased COL1A1 expression (+116%) and cell proliferation (+10.9%). These effects were attenuated by methyllycaconitine citrate, indicating the profibrotic effects of nicotine via α7nAChR. CONCLUSIONS: Nicotine aggravates liver fibrosis induced by other factors by activating α7nAChR on HSCs, thereby increasing their collagen-producing capacity. We suggest the profibrotic effect of nicotine is mediated through α7nAChRs.


Asunto(s)
Tetracloruro de Carbono , Cadena alfa 1 del Colágeno Tipo I , Colágeno Tipo I , Células Estrelladas Hepáticas , Cirrosis Hepática , Nicotina , Receptor Nicotínico de Acetilcolina alfa 7 , Animales , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/genética , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/efectos de los fármacos , Nicotina/efectos adversos , Ratones , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Humanos , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I/metabolismo , Ratas , Masculino , Proliferación Celular/efectos de los fármacos , Aconitina/farmacología , Aconitina/análogos & derivados , Línea Celular , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta1/metabolismo , Ratones Noqueados , Agonistas Nicotínicos/farmacología
5.
J Vet Med Sci ; 86(7): 824-827, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38839347

RESUMEN

The expression of nicotinic acetylcholine receptor (nAChR) subunits on various immune cells suggests their involvement in allergic rhinitis. However, how exactly they contribute to this pathogenesis is not yet confirmed. Our present study examined the therapeutic potential of GTS-21, an α7 nAChR agonist, for treating allergic rhinitis by employing its mouse models. GTS-21 treatment reduced allergen-induced immediate nasal response in ovalbumin (OVA)-sensitized model. However, nasal hyperresponsiveness or eosinophil infiltration elicited in either the OVA-sensitized or T helper 2 cell-transplanted model was not affected by GTS-21. GTS-21 did not alter allergen-induced passive cutaneous anaphylaxis response in anti-dinitrophenyl IgE-sensitized mice. This evidence implies GTS-21's potential to alleviate allergic rhinitis without perturbing T cells or mast cells.


Asunto(s)
Alérgenos , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , Ovalbúmina , Rinitis Alérgica , Receptor Nicotínico de Acetilcolina alfa 7 , Animales , Rinitis Alérgica/tratamiento farmacológico , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Femenino , Ratones , Piridinas/farmacología , Piridinas/uso terapéutico , Agonistas Nicotínicos/uso terapéutico , Agonistas Nicotínicos/farmacología , Compuestos de Bencilideno/farmacología , Compuestos de Bencilideno/uso terapéutico
6.
Biochem Pharmacol ; 225: 116263, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38735444

RESUMEN

Although α2 was the first neuronal nicotinic acetylcholine receptor (nAChR) receptor subunit to be cloned, due to its low level of expression in rodent brain, its study has largely been neglected. This study provides a comparison of the α2 and α4 structures and their functional similarities, especially in regard to the existence of low and high sensitivity forms based on subunit stoichiometry. We show that the pharmacological profiles of the low and high sensitivity forms of α2ß2 and α4ß2 receptors are very similar in their responses to nicotine, with high sensitivity receptors showing protracted responses. Sazetidine A, an agonist that is selective for the high sensitivity α4 receptors also selectively activates high sensitivity α2 receptors. Likewise, α2 receptors have similar responses as α4 receptors to the positive allosteric modulators (PAMs) desformylflustrabromine (dFBr) and NS9283. We show that the partial agonists for α4ß2 receptors, cytisine and varenicline are also partial agonists for α2ß2 receptors. Studies have shown that levels of α2 expression may be much higher in the brains of primates than those of rodents, suggesting a potential importance for human therapeutics. High-affinity nAChR have been studied in humans with PET ligands such as flubatine. We show that flubatine has similar activity with α2ß2 and α4ß2 receptors so that α2 receptors will also be detected in PET studies that have previously presumed to selectively detect α4ß2 receptors. Therefore, α2 receptors need more consideration in the development of therapeutics to manage nicotine addiction and declining cholinergic function in age and disease.


Asunto(s)
Agonistas Nicotínicos , Receptores Nicotínicos , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/genética , Animales , Agonistas Nicotínicos/farmacología , Humanos , Nicotina/farmacología , Nicotina/metabolismo , Xenopus laevis , Azetidinas/farmacología , Quinolizinas/farmacología , Vareniclina/farmacología , Azocinas/farmacología , Alcaloides de Quinolizidina , Piridinas
7.
J Clin Psychopharmacol ; 44(4): 362-368, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38752924

RESUMEN

PURPOSE/BACKGROUND: People who smoke cigarettes and drink alcohol heavily are less likely to quit smoking compared with those who do not drink heavily. The current study examined the effects of a 12-week treatment phase of combination varenicline and nicotine patch compared with placebo and nicotine patch on smoking cessation (primary outcome) and alcohol consumption (secondary outcome) in heavy drinking smokers at 26-week follow-up. METHODS/PROCEDURES: Participants were daily smokers who met heavy drinking criteria. They were randomly assigned to receive either varenicline and nicotine patch (n = 61) or placebo and nicotine patch (n = 61) for 12 weeks. At week 26, self-reports of point prevalence cigarette abstinence were biochemically confirmed, and past-month alcohol drinking days and heavy drinking days were assessed. FINDINGS/RESULTS: At week 26, smoking quit rates did not differ by treatment group (25% varenicline and 26% placebo). Relative to week 12 outcomes, week 26 quit rates significantly dropped off in the varenicline group but not in the placebo group. Alcohol drinking reductions for the whole sample that were previously observed from baseline to week 12 were sustained at week 26, although they did not differ between treatment groups. IMPLICATIONS/CONCLUSIONS: In heavy drinking smokers, smoking cessation success was evident in a quarter of the total sample at 3 months postmedication discontinuation. At this time, quit rates were the same between those who received varenicline and nicotine patch and those who received nicotine patch alone. Future research is warranted to examine what may aid in longer-term smoking quit rates in heavy drinking smokers.


Asunto(s)
Consumo de Bebidas Alcohólicas , Agentes para el Cese del Hábito de Fumar , Cese del Hábito de Fumar , Dispositivos para Dejar de Fumar Tabaco , Vareniclina , Humanos , Vareniclina/administración & dosificación , Vareniclina/farmacología , Cese del Hábito de Fumar/métodos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Estudios de Seguimiento , Agentes para el Cese del Hábito de Fumar/administración & dosificación , Quimioterapia Combinada , Resultado del Tratamiento , Agonistas Nicotínicos/administración & dosificación , Método Doble Ciego
8.
J Med Chem ; 67(11): 8642-8666, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38748608

RESUMEN

There is an urgent need for nonopioid treatments for chronic and neuropathic pain to provide effective alternatives amid the escalating opioid crisis. This study introduces novel compounds targeting the α9 nicotinic acetylcholine receptor (nAChR) subunit, which is crucial for pain regulation, inflammation, and inner ear functions. Specifically, it identifies novel substituted carbamoyl/amido/heteroaryl dialkylpiperazinium iodides as potent agonists selective for human α9 and α9α10 over α7 nAChRs, particularly compounds 3f, 3h, and 3j. Compound 3h (GAT2711) demonstrated a 230 nM potency as a full agonist at α9 nAChRs, being 340-fold selective over α7. Compound 3c was 10-fold selective for α9α10 over α9 nAChR. Compounds 2, 3f, and 3h inhibited ATP-induced interleukin-1ß release in THP-1 cells. The analgesic activity of 3h was fully retained in α7 knockout mice, suggesting that analgesic effects were potentially mediated through α9* nAChRs. Our findings provide a blueprint for developing α9*-specific therapeutics for pain.


Asunto(s)
Analgésicos , Inflamación , Piperazinas , Receptores Nicotínicos , Animales , Humanos , Masculino , Ratones , Analgésicos/farmacología , Analgésicos/química , Analgésicos/síntesis química , Analgésicos/uso terapéutico , Inflamación/tratamiento farmacológico , Ratones Noqueados , Agonistas Nicotínicos/farmacología , Agonistas Nicotínicos/química , Agonistas Nicotínicos/uso terapéutico , Agonistas Nicotínicos/síntesis química , Dolor/tratamiento farmacológico , Piperazinas/farmacología , Piperazinas/química , Piperazinas/síntesis química , Piperazinas/uso terapéutico , Receptores Nicotínicos/metabolismo , Sales (Química)/química , Sales (Química)/farmacología , Relación Estructura-Actividad , Yoduros/química
9.
Neuropharmacology ; 255: 110001, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38750804

RESUMEN

Emerging evidence suggests an important role of astrocytes in mediating behavioral and molecular effects of commonly misused drugs. Passive exposure to nicotine alters molecular, morphological, and functional properties of astrocytes. However, a potential involvement of astrocytes in nicotine reinforcement remains largely unexplored. The overall hypothesis tested in the current study is that astrocytes play a critical role in nicotine reinforcement. Protein levels of the astrocyte marker glial fibrillary acidic protein (GFAP) were examined in key mesocorticolimbic regions following chronic nicotine intravenous self-administration. Fluorocitrate, a metabolic inhibitor of astrocytes, was tested for its effects on behaviors related to nicotine reinforcement and relapse. Effects of fluorocitrate on extracellular neurotransmitter levels, including glutamate, GABA, and dopamine, were determined with microdialysis. Chronic nicotine intravenous self-administration increased GFAP expression in the nucleus accumbens core (NACcr), but not other key mesocorticolimbic regions, compared to saline intravenous self-administration. Both intra-ventricular and intra-NACcr microinjection of fluorocitrate decreased nicotine self-administration. Intra-NACcr fluorocitrate microinjection also inhibited cue-induced reinstatement of nicotine seeking. Local perfusion of fluorocitrate decreased extracellular glutamate levels, elevated extracellular dopamine levels, but did not alter extracellular GABA levels in the NACcr. Fluorocitrate did not alter basal locomotor activity. These results indicate that nicotine reinforcement upregulates the astrocyte marker GFAP expression in the NACcr, metabolic inhibition of astrocytes attenuates nicotine reinforcement and relapse, and metabolic inhibition of astrocytes disrupts extracellular dopamine and glutamate transmission. Overall, these findings suggest that astrocytes play an important role in nicotine reinforcement and relapse, potentially through regulation of extracellular glutamate and dopamine neurotransmission.


Asunto(s)
Astrocitos , Citratos , Dopamina , Ácido Glutámico , Nicotina , Núcleo Accumbens , Ratas Wistar , Autoadministración , Animales , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Nicotina/farmacología , Nicotina/administración & dosificación , Masculino , Ácido Glutámico/metabolismo , Dopamina/metabolismo , Citratos/farmacología , Citratos/administración & dosificación , Ratas , Proteína Ácida Fibrilar de la Glía/metabolismo , Agonistas Nicotínicos/farmacología , Agonistas Nicotínicos/administración & dosificación , Microdiálisis , Refuerzo en Psicología , Ácido gamma-Aminobutírico/metabolismo
10.
JAMA ; 331(20): 1722-1731, 2024 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-38696203

RESUMEN

Importance: Most people who smoke do not quit on their initial attempt. Objective: To determine the best subsequent strategy for nonabstinence following initial treatment with varenicline or combined nicotine replacement therapy (CNRT). Design, Setting, and Participants: Using a double-blind, placebo-controlled, sequential multiple assignment randomized trial, 490 volunteers were randomized to receive 6 weeks of varenicline or CNRT. After 6 weeks, nonabstainers were rerandomized to continue, switch, or increase medication dosage for 6 additional weeks. The study was conducted from June 2015 through October 2019 in a Texas tobacco treatment clinic. Interventions: The initial treatment was 2 mg/d of varenicline or the combined replacement therapy of a 21-mg patch plus 2-mg lozenge. The rerandomized participants either continued with their initial therapies, switched between varenicline and CNRT, or increased dosages either to 3-mg or more of varenicline or to a 42-mg patch and lozenges. All received weekly brief counseling. Main Outcomes and Measures: Biochemically verified 7-day point prevalence abstinence at the end of treatment at 12 weeks. Results: The 490 randomized participants (210 female [43%], 287 non-Hispanic White [58%], mean age, 48.1 years) smoked an average of 20 cigarettes per day. After the first phase, 54 participants in the CNRT group were abstinent and continued their therapy; of the 191 who were not abstinent, 151 were rerandomized, and the 40 who did not return for rerandomization were assigned to continue their initial CNRT condition in phase 2. The end-of-treatment abstinence rate for the 191 phase 1 nonabstainers was 8% (95% credible interval [CrI], 6% to 10%) for the 90 (47%) who continued at the dosage condition, 14% (CrI, 10% to 18%) for the 50 (33%) who increased their dosage, and 14% (95% CrI, 10% to 18%) for the 51 (34%) who switched to varenicline (absolute risk difference [RD], 6%; 95% CrI, 6% to 11%) with more than 99% posterior probability that either strategy conferred benefit over continuing the initial dosage. After the first phase, 88 participants in the varenicline group were abstinent and continued their therapy; of the 157 who were not abstinent, 122 were rerandomized and 35 who did not return for rerandomization were assigned to continue with the varenicline condition. The end-of-treatment abstinence rate for the 157 phase 1 nonabstainers was 20% (95% CrI, 16% to 26%) for the 39 (32%) who increased their varenicline dosage, 0 (95% CrI, 0 to 0) for the 41 (34%) who switched CNRT, and 3% (95% CrI, 1% to 4%) for the 77 (49%) who were assigned to the continued varenicline condition (absolute RD, -3%; 95% CrI, -4% to -1%) with more than 99% posterior probability that continuing varenicline at the initial dosage was worse than switching to a higher dosage. Furthermore, increasing the varenicline dosage had an absolute RD of 18% (95% CrI, 13% to 24%) and a more than 99% posterior probability of conferring benefit. The secondary outcome of continuous abstinence at 6 months indicated that only increased dosages of the CNRT and varenicline provided benefit over continuation of the initial treatment dosages. Conclusions and Relevance: For individuals who smoked but did not achieve abstinence after treatment with varenicline, increasing the dosage enhanced abstinence vs continuing, whereas for nonabstainers initially treated with CNRT, a dosage increase or switch to varenicline enhanced abstinence and may be viable rescue strategies. Trial Registration: ClinicalTrials.gov Identifier: NCT02271919.


Asunto(s)
Nicotina , Agonistas Nicotínicos , Agentes para el Cese del Hábito de Fumar , Cese del Hábito de Fumar , Vareniclina , Femenino , Humanos , Masculino , Persona de Mediana Edad , Método Doble Ciego , Nicotina/administración & dosificación , Nicotina/efectos adversos , Nicotina/uso terapéutico , Agonistas Nicotínicos/administración & dosificación , Agonistas Nicotínicos/uso terapéutico , Cese del Hábito de Fumar/métodos , Agentes para el Cese del Hábito de Fumar/uso terapéutico , Agentes para el Cese del Hábito de Fumar/efectos adversos , Agentes para el Cese del Hábito de Fumar/administración & dosificación , Insuficiencia del Tratamiento , Vareniclina/uso terapéutico , Vareniclina/administración & dosificación , Vareniclina/efectos adversos , Blanco
13.
Artículo en Inglés | MEDLINE | ID: mdl-38795824

RESUMEN

As one of the leading causes of death and serious illnesses, tobacco smoking remains a significant issue in modern societies. Many individuals smoke during adolescence, a trend that has been exacerbated by the prevalence of vaping among young people. In this context, studying the behavioral effects induced by nicotine administration in male and female rats, during the adolescent period, assumes great importance because it can help to better understand the dynamics underlying tobacco use in the two sexes. For this purpose, we employed 4 groups of rats, 2 male and 2 female groups, chronically treated with saline or nicotine 3 mg/kg i.p. for 30 days, spanning from postnatal day 30 to postnatal day 60. Utilizing quantitative analyses and T-pattern detection and analysis, our findings revealed a complex and multifaceted behavioral reorganization in adolescent rats subjected to chronic nicotine administration. Specifically, we observed an increase of anxiety in males and a reduction in females. The distinctive structural changes, induced by chronic nicotine in both sexes, have significant implications, from a translational perspective, for studies on nicotine dependence disorders.


Asunto(s)
Nicotina , Animales , Nicotina/farmacología , Nicotina/efectos adversos , Femenino , Masculino , Ratas , Caracteres Sexuales , Agonistas Nicotínicos/farmacología , Conducta Animal/efectos de los fármacos , Ansiedad/inducido químicamente , Ratas Wistar , Factores Sexuales
14.
ACS Chem Neurosci ; 15(9): 1738-1754, 2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38613458

RESUMEN

Iboga alkaloids, also known as coronaridine congeners, have shown promise in the treatment of alcohol and opioid use disorders. The objective of this study was to evaluate the effects of catharanthine and 18-methoxycoronaridine (18-MC) on dopamine (DA) transmission and cholinergic interneurons in the mesolimbic DA system, nicotine-induced locomotor activity, and nicotine-taking behavior. Utilizing ex vivo fast-scan cyclic voltammetry (FSCV) in the nucleus accumbens core of male mice, we found that catharanthine or 18-MC differentially inhibited evoked DA release. Catharanthine inhibition of evoked DA release was significantly reduced by both α4 and α6 nicotinic acetylcholine receptors (nAChRs) antagonists. Additionally, catharanthine substantially increased DA release more than vehicle during high-frequency stimulation, although less potently than an α4 nAChR antagonist, which confirms previous work with nAChR antagonists. Interestingly, while catharanthine slowed DA reuptake measured via FSCV ex vivo, it also increased extracellular DA in striatal dialysate from anesthetized mice in vivo in a dose-dependent manner. Superfusion of catharanthine or 18-MC inhibited the firing rate of striatal cholinergic interneurons in a concentration dependent manner, which are known to potently modulate presynaptic DA release. Catharanthine or 18-MC suppressed acetylcholine currents in oocytes expressing recombinant rat α6/α3ß2ß3 or α6/α3ß4 nAChRs. In behavioral experiments using male Sprague-Dawley rats, systemic administration of catharanthine or 18-MC blocked nicotine enhancement of locomotor activity. Importantly, catharanthine attenuated nicotine self-administration in a dose-dependent manner while having no effect on food reinforcement. Lastly, administration of catharanthine and nicotine together greatly increased head twitch responses, indicating a potential synergistic hallucinogenic effect. These findings demonstrate that catharanthine and 18-MC have similar, but not identical effects on striatal DA dynamics, striatal cholinergic interneuron activity and nicotine psychomotor effects.


Asunto(s)
Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Dopamina , Ibogaína , Ibogaína/análogos & derivados , Nicotina , Receptores Nicotínicos , Animales , Dopamina/metabolismo , Masculino , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/efectos de los fármacos , Nicotina/farmacología , Ibogaína/farmacología , Ratones , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Ratones Endogámicos C57BL , Antagonistas Nicotínicos/farmacología , Oocitos/efectos de los fármacos , Agonistas Nicotínicos/farmacología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Autoadministración , Xenopus laevis , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Relación Dosis-Respuesta a Droga , Actividad Motora/efectos de los fármacos
15.
High Blood Press Cardiovasc Prev ; 31(3): 225-237, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38668958

RESUMEN

INTRODUCTION: Smoke from traditional cigarettes and e-cigarette aerosols have distinct chemical compositions that may impact blood pressure (BP) and heart rate (HR) differently. AIMS: This study compared the impact of nicotine-containing e-cigarettes (EC+) versus nicotine-free (EC-) on BP, HR and endothelial markers, and assessed if EC+ posed fewer risks than tobacco cigarettes (TC). METHODS: Electronic databases were searched from inception until November 2023 for studies reporting changes in systolic and diastolic BP (SBP, DBP) and HR and endothelial parameters before and after the use of EC+, EC- and TC. Data were analyzed using weighted mean differences (WMDs) and 95% confidence intervals (CIs). RESULTS: Fifteen studies (n = 752) were included in our meta-analysis. We demonstrate that EC+ significantly increased systolic BP (WMD = 3.41, 95% CI [0.1,6.73], p = 0.04], diastolic BP (WMD = 3.42, 95% CI [1.75, 5.09]; p < 0.01], and HR (WMD = 5.36 BPM, 95% CI [1.87, 8.85]; p < 0.01) compared to EC-. However, EC+ was observed to cause less detrimental effect on SBP (WMD = - 4.72 mmHg, 95% CI [- 6.58, - 2.86], p < 0.01), and HR (WMD = - 3.11 BPM, 95% CI [- 4.54, - 1.68]; p < 0.01) as compared to TC with no difference on DBP (WMD = - 1.14 mmHg, 95% CI [- 2.38, 0.1]; p = 0.07). EC+ also led to greater deterioration of endothelial parameters as compared to EC- but to a lesser degree as compared to TC. CONCLUSION: EC+ shows greater impairment in hemodynamic and endothelial parameters than EC- but less than TC. Additional studies are needed to evaluate prolonged effects of EC use.


Asunto(s)
Presión Sanguínea , Sistemas Electrónicos de Liberación de Nicotina , Endotelio Vascular , Frecuencia Cardíaca , Nicotina , Productos de Tabaco , Vapeo , Humanos , Vapeo/efectos adversos , Presión Sanguínea/efectos de los fármacos , Endotelio Vascular/fisiopatología , Endotelio Vascular/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Nicotina/efectos adversos , Nicotina/administración & dosificación , Masculino , Femenino , Productos de Tabaco/efectos adversos , Adulto , Persona de Mediana Edad , Agonistas Nicotínicos/efectos adversos , Agonistas Nicotínicos/administración & dosificación , Medición de Riesgo , Factores de Riesgo , Adulto Joven , Anciano , Hemodinámica/efectos de los fármacos , Factores de Tiempo
16.
Behav Brain Res ; 467: 115019, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38677331

RESUMEN

Nicotine smoking contributes to many preventable disabilities, diseases and deaths. Targeting nicotine reward and withdrawal is a basis for the majority of smoking cessation pharmacotherapies. Due to the emergence of interest in 5-HT2A receptor modulators for numerous psychiatric disorders, we investigated the effect of nelotanserin, a 5-HT2A receptor inverse agonist, on nicotine reward and withdrawal in ICR mice. In nicotine-dependent mice, nelotanserin dose-dependently reduced somatic signs of nicotine withdrawal and thermal hyperalgesia as measured in the hot plate test. However, nelotanserin had no effect on anxiety-like behavior and failed to reduce nicotine reward as measured in the conditioned place preference test. Our results suggest that inverse agonism of the 5-HT2A receptor may be a feasible novel mechanism for smoking cessation by reducing both physical withdrawal and thermal hyperalgesia associated with nicotine abstinence but may require complementary pharmacotherapies targeting affective and reward-associated decrements to improve cessation outcomes.


Asunto(s)
Ratones Endogámicos ICR , Nicotina , Recompensa , Agonistas del Receptor de Serotonina 5-HT2 , Síndrome de Abstinencia a Sustancias , Animales , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Nicotina/farmacología , Nicotina/administración & dosificación , Masculino , Agonistas del Receptor de Serotonina 5-HT2/farmacología , Ratones , Relación Dosis-Respuesta a Droga , Tabaquismo/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/inducido químicamente , Receptor de Serotonina 5-HT2A/metabolismo , Receptor de Serotonina 5-HT2A/efectos de los fármacos , Ansiedad/tratamiento farmacológico , Agonistas Nicotínicos/farmacología , Agonistas Nicotínicos/administración & dosificación
17.
Neuropharmacology ; 253: 109959, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38648925

RESUMEN

Nicotine use produces psychoactive effects, and chronic use is associated with physiological and psychological symptoms of addiction. However, chronic nicotine use is known to decrease food intake and body weight gain, suggesting that nicotine also affects central metabolic and appetite regulation. We recently showed that acute nicotine self-administration in nicotine-dependent animals produces a short-term increase in food intake, contrary to its long-term decrease of feeding behavior. As feeding behavior is regulated by complex neural signaling mechanisms, this study aimed to test the hypothesis that nicotine intake in animals exposed to chronic nicotine may increase activation of pro-feeding regions and decrease activation of pro-satiety regions to produce the acute increase in feeding behavior. FOS immunohistochemistry revealed that acute nicotine intake in nicotine self-administering animals increased activation of the pro-feeding arcuate and lateral hypothalamic nuclei and decreased activation of the pro-satiety parabrachial nucleus. Regional correlational analysis also showed that acute nicotine changes the functional connectivity of the hunger/satiety network. Further dissection of the role of the arcuate nucleus using electrophysiology found that putative POMC neurons in animals given chronic nicotine exhibited decreased firing following acute nicotine application. These brain-wide central signaling changes may contribute to the acute increase in feeding behavior we see in rats after acute nicotine and provide new areas of focus for studying both nicotine addiction and metabolic regulation.


Asunto(s)
Encéfalo , Nicotina , Animales , Nicotina/farmacología , Masculino , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ratas , Ratas Sprague-Dawley , Agonistas Nicotínicos/farmacología , Conducta Alimentaria/efectos de los fármacos , Proopiomelanocortina/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Autoadministración , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Anorexia/inducido químicamente
18.
Int J Mol Sci ; 25(8)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38674000

RESUMEN

Stimulation of the alpha 7 nicotinic acetylcholine receptor (α7nAChR) has shown beneficial effects in several acute inflammatory disease models. This study aims to examine whether treatment with the selective α7nAChR agonist PHA 568487 can dampen inflammation and thereby improve cardiac function after myocardial infarction in mice. The possible anti-inflammatory properties of α7nAChR agonist PHA 568487 were tested in vivo using the air pouch model and in a permanent occlusion model of acute myocardial infarction in mice. Hematologic parameters and cytokine levels were determined. Infarct size and cardiac function were assessed via echocardiography 24 h and one week after the infarction. Treatment with α7nAChR agonist PHA 568487 decreased 12 (CCL27, CXCL5, IL6, CXCL10, CXCL11, CXCL1, CCL2, MIP1a, MIP2, CXCL16, CXCL12 and CCL25) out of 33 cytokines in the air pouch model of acute inflammation. However, α7nAChR agonist PHA 568487 did not alter infarct size, ejection fraction, cardiac output or stroke volume at 24 h or at 7 days after the myocardial infarction compared with control mice. In conclusion, despite promising immunomodulatory effects in the acute inflammatory air pouch model, α7nAChR agonist PHA 568487 did not affect infarct size or cardiac function after a permanent occlusion model of acute myocardial infarction in mice. Consequently, this study does not strengthen the hypothesis that stimulation of the α7nAChR is a future treatment strategy for acute myocardial infarction when reperfusion is lacking. However, whether other agonists of the α7nAChR can have different effects remains to be investigated.


Asunto(s)
Modelos Animales de Enfermedad , Inflamación , Infarto del Miocardio , Receptor Nicotínico de Acetilcolina alfa 7 , Animales , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Ratones , Inflamación/tratamiento farmacológico , Inflamación/patología , Inflamación/metabolismo , Masculino , Citocinas/metabolismo , Agonistas Nicotínicos/farmacología , Agonistas Nicotínicos/uso terapéutico , Piridinas/farmacología , Piridinas/uso terapéutico , Ratones Endogámicos C57BL , Quinuclidinas/farmacología , Quinuclidinas/uso terapéutico , Bencilaminas/farmacología , Bencilaminas/uso terapéutico , Compuestos de Bencilideno/farmacología
19.
Psychiatry Res ; 335: 115874, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38564922

RESUMEN

Smoking cessation medications have the potential to affect the functioning of the nervous system, leading to sleep disturbances. Our study aimed to compare the sleep-related side effects (such as insomnia, abnormal dreams, nightmares, and somnolence) induced by different smoking cessation medications in non-psychiatric smokers. We conducted a thorough search of five electronic databases (Cochrane, EMBASE, PubMed, PsycInfo, and Web of Science) for randomized controlled trials. This study was registered with the PROSPERO (registration number CRD42022347976). A total of 79 full-text articles, encompassing 36,731 participants, were included in our analysis. Individuals using bupropion, bupropion in combination with a nicotinic acetylcholine receptor agonist (NRA), and bupropion in conjunction with nicotine replacement therapy (NRT) exhibited a higher likelihood of experiencing insomnia compared to those using NRT alone. Bupropion plus NRA had the highest ranking on the surface under the cumulative ranking curve (SUCRA) for insomnia risk, while placebo had the lowest ranking. Additionally, NRA plus NRT ranked first for abnormal dream outcomes, NRA alone for nightmares, and nortriptyline for somnolence, based on the SUCRA results. Healthcare providers should exercise caution when prescribing smoking cessation drugs, particularly in consideration of their potential sleep-related side effects.


Asunto(s)
Trastornos del Inicio y del Mantenimiento del Sueño , Cese del Hábito de Fumar , Humanos , Cese del Hábito de Fumar/psicología , Bupropión/efectos adversos , Vareniclina/uso terapéutico , Fumar/psicología , Metaanálisis en Red , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Somnolencia , Dispositivos para Dejar de Fumar Tabaco/efectos adversos , Ensayos Clínicos Controlados Aleatorios como Asunto , Agonistas Nicotínicos/efectos adversos , Sueño
20.
Neuropsychopharmacology ; 49(7): 1171-1182, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38521861

RESUMEN

The majority of lifetime smokers begin using nicotine during adolescence, a critical period of brain development wherein neural circuits critical for mood, affect and cognition are vulnerable to drug-related insults. Specifically, brain regions such as the medial prefrontal cortex (mPFC), the ventral tegmental area (VTA), nucleus accumbens (NAc) and hippocampus, are implicated in both nicotine dependence and pathological phenotypes linked to mood and anxiety disorders. Clinical studies report that females experience higher rates of mood/anxiety disorders and are more resistant to smoking cessation therapies, suggesting potential sex-specific responses to nicotine exposure and later-life neuropsychiatric risk. However, the potential neural and molecular mechanisms underlying such sex differences are not clear. In the present study, we compared the impacts of adolescent nicotine exposure in male vs. female rat cohorts. We performed a combination of behavioral, electrophysiological and targeted protein expression analyses along with matrix assisted laser deionization imaging (MALDI) immediately post-adolescent exposure and later in early adulthood. We report that adolescent nicotine exposure induced long-lasting anxiety/depressive-like behaviors, disrupted neuronal activity patterns in the mPFC-VTA network and molecular alterations in various neural regions linked to affect, anxiety and cognition. Remarkably, these phenotypes were only observed in males and/or were expressed in the opposite direction in females. These findings identify a series of novel, sex-selective biomarkers for adolescent nicotine-induced neuropsychiatric risk, persisting into adulthood.


Asunto(s)
Ansiedad , Nicotina , Caracteres Sexuales , Animales , Masculino , Femenino , Nicotina/toxicidad , Nicotina/efectos adversos , Ansiedad/inducido químicamente , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Fenotipo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas Sprague-Dawley , Agonistas Nicotínicos/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA