Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 319
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 326(6): H1337-H1349, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38551482

RESUMEN

Nicotine is the primary addictive component of tobacco products. Through its actions on the heart and autonomic nervous system, nicotine exposure is associated with electrophysiological changes and increased arrhythmia susceptibility. To assess the underlying mechanisms, we treated rabbits with transdermal nicotine (NIC, 21 mg/day) or control (CT) patches for 28 days before performing dual optical mapping of transmembrane potential (RH237) and intracellular Ca2+ (Rhod-2 AM) in isolated hearts with intact sympathetic innervation. Sympathetic nerve stimulation (SNS) was performed at the first to third thoracic vertebrae, and ß-adrenergic responsiveness was additionally evaluated following norepinephrine (NE) perfusion. Baseline ex vivo heart rate (HR) and SNS stimulation threshold were higher in NIC versus CT (P = 0.004 and P = 0.003, respectively). Action potential duration alternans emerged at longer pacing cycle lengths (PCL) in NIC versus CT at baseline (P = 0.002) and during SNS (P = 0.0003), with similar results obtained for Ca2+ transient alternans. SNS shortened the PCL at which alternans emerged in CT but not in NIC hearts. NIC-exposed hearts tended to have slower and reduced HR responses to NE perfusion, but ventricular responses to NE were comparable between groups. Although fibrosis was unaltered, NIC hearts had lower sympathetic nerve density (P = 0.03) but no difference in NE content versus CT. These results suggest both sympathetic hypoinnervation of the myocardium and regional differences in ß-adrenergic responsiveness with NIC. This autonomic remodeling may contribute to the increased risk of arrhythmias associated with nicotine exposure, which may be further exacerbated with long-term use.NEW & NOTEWORTHY Here, we show that chronic nicotine exposure was associated with increased heart rate, increased susceptibility to alternans, and reduced sympathetic electrophysiological responses in the intact rabbit heart. We suggest that this was due to sympathetic hypoinnervation of the myocardium and diminished ß-adrenergic responsiveness of the sinoatrial node following nicotine treatment. Though these differences did not result in increased arrhythmia propensity in our study, we hypothesize that prolonged nicotine exposure may exacerbate this proarrhythmic remodeling.


Asunto(s)
Potenciales de Acción , Frecuencia Cardíaca , Corazón , Nicotina , Sistema Nervioso Simpático , Animales , Nicotina/toxicidad , Nicotina/efectos adversos , Conejos , Frecuencia Cardíaca/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Corazón/inervación , Corazón/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología , Masculino , Agonistas Nicotínicos/toxicidad , Agonistas Nicotínicos/administración & dosificación , Señalización del Calcio/efectos de los fármacos , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/metabolismo , Parche Transdérmico , Preparación de Corazón Aislado , Administración Cutánea , Norepinefrina/metabolismo
2.
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
3.
Cardiovasc Toxicol ; 24(5): 435-471, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38555547

RESUMEN

Cigarette smoking is positively and robustly associated with cardiovascular disease (CVD), including hypertension, atherosclerosis, cardiac arrhythmias, stroke, thromboembolism, myocardial infarctions, and heart failure. However, after more than a decade of ENDS presence in the U.S. marketplace, uncertainty persists regarding the long-term health consequences of ENDS use for CVD. New approach methods (NAMs) in the field of toxicology are being developed to enhance rapid prediction of human health hazards. Recent technical advances can now consider impact of biological factors such as sex and race/ethnicity, permitting application of NAMs findings to health equity and environmental justice issues. This has been the case for hazard assessments of drugs and environmental chemicals in areas such as cardiovascular, respiratory, and developmental toxicity. Despite these advances, a shortage of widely accepted methodologies to predict the impact of ENDS use on human health slows the application of regulatory oversight and the protection of public health. Minimizing the time between the emergence of risk (e.g., ENDS use) and the administration of well-founded regulatory policy requires thoughtful consideration of the currently available sources of data, their applicability to the prediction of health outcomes, and whether these available data streams are enough to support an actionable decision. This challenge forms the basis of this white paper on how best to reveal potential toxicities of ENDS use in the human cardiovascular system-a primary target of conventional tobacco smoking. We identify current approaches used to evaluate the impacts of tobacco on cardiovascular health, in particular emerging techniques that replace, reduce, and refine slower and more costly animal models with NAMs platforms that can be applied to tobacco regulatory science. The limitations of these emerging platforms are addressed, and systems biology approaches to close the knowledge gap between traditional models and NAMs are proposed. It is hoped that these suggestions and their adoption within the greater scientific community will result in fresh data streams that will support and enhance the scientific evaluation and subsequent decision-making of tobacco regulatory agencies worldwide.


Asunto(s)
Enfermedades Cardiovasculares , Sistemas Electrónicos de Liberación de Nicotina , Vapeo , Humanos , Medición de Riesgo , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/prevención & control , Animales , Vapeo/efectos adversos , Vapeo/tendencias , Factores de Riesgo , Nicotina/efectos adversos , Nicotina/administración & dosificación , Agonistas Nicotínicos/efectos adversos , Agonistas Nicotínicos/administración & dosificación , Agonistas Nicotínicos/toxicidad , Seguridad de Productos para el Consumidor , Sistema Cardiovascular/efectos de los fármacos , Cardiotoxicidad , Factores de Riesgo de Enfermedad Cardiaca , Cigarrillo Electrónico a Vapor/efectos adversos
4.
Neurosci Lett ; 823: 137654, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38281695

RESUMEN

The α7 neuronal nicotinic acetylcholine receptor (α7 nAChR) is a potential target for the development of Parkinson's disease (PD) therapeutics. α-Synuclein (α-Syn), a principal component of Lewy bodies (cytoplasmic inclusions), is a major contributor to PD pathophysiology. Previous studies have demonstrated that activating α7 nAChR protects against nigrostriatal dopamine degeneration in acute and chronic PD animal models induced by 6-hydroxydopamine and rotenone, respectively. In the present study, we investigated the effects of PNU282987, a selective α7 nAChR agonist, against α-Syn-induced neurotoxicity in α-SynWT-, α-SynA30P-, and α-SynE46K-N2a cells. PNU282987 exhibited substantial neuroprotection against both wild-type and mutant-type α-Syn-induced toxicity. Furthermore, PNU282987 promoted transcription factor EB activity and reduced intracellular α-Syn protein levels through autophagy induction. These results highlight the therapeutic potential of α7 nAChR activation in diseases characterized by α-Syn aggregation, such as PD.


Asunto(s)
Compuestos Bicíclicos con Puentes , Síndromes de Neurotoxicidad , Enfermedad de Parkinson , Receptores Nicotínicos , Animales , alfa-Sinucleína/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7 , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Benzamidas/farmacología , Agonistas Nicotínicos/toxicidad , Receptores Nicotínicos/metabolismo
5.
J Neuroimmune Pharmacol ; 18(3): 413-426, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37351737

RESUMEN

Perinatal exposure to prescription opioids pose a critical public health risk. Notably, research has found significant neurodevelopmental and behavioral deficits between in utero (IUO) and postnatal (PNO) oxycodone-exposed offspring but there is a notable gap in knowledge regarding the interaction of these groups to other drug exposure, particularly nicotine exposure. Nicotine's widespread use represents a ubiquitous clinical interaction that current research does not address. Children often experiment with drugs and risky behavior; therefore, adolescence is a key timepoint to characterize. This study employed an integrated systems approach to investigate escalating nicotine exposure in adolescence and subsequent nicotine withdrawal in the IUO- and PNO-offspring. Western blot analysis found synaptic protein alterations, especially upregulation of synaptophysin in IUO-withdrawal animals. RT-qPCR further validated immune dysfunction in the central nervous system (CNS). Peripheral nicotine metabolism was consistent with increased catabolism of nicotine concerning IUO animals. Lastly, behavioral assays found subtle deficits to withdrawal in nociception and anxiety-like behavior. This study showed, for the first time, the vulnerabilities of PNO- and IUO-exposed groups concerning nicotine use during early adolescence and withdrawal. Graphical Abstract.


Asunto(s)
Nicotina , Agonistas Nicotínicos , Embarazo , Animales , Femenino , Niño , Humanos , Adolescente , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Oxicodona/efectos adversos
6.
Int J Mol Sci ; 23(3)2022 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-35163155

RESUMEN

While the cognitive enhancing effects of nicotine use have been well documented, it has also been shown to impair decision making. The goal of this study was to determine if exposure to nicotine vapor increases risky decision making. The study also aims to investigate possible long-term effects of nicotine vapor exposure on the expression of genes coding for cholinergic and dopaminergic receptors in brain. Thirty-two adult male Sprague Dawley rats were exposed to 24 mg/mL nicotine vapor or vehicle control, immediately followed by testing in the probability discounting task for 10 consecutive days. Fifty-four days after the 10-day vapor exposure, animals were sacrificed and expression of genes coding for the α4 and ß2 cholinergic receptor subunits, and dopamine D1 and D2 receptors, were analyzed using RT-PCR. Exposure to nicotine vapor caused an immediate and transient increase in risky choice. Analyses of gene expression identified significant reductions in CHRNB2 and DRD1 in the nucleus accumbens core and CHRNB2 and DRD2 in the medial prefrontal cortex of rats previously exposed to nicotine vapor, relative to vehicle controls. Results provide data on the negative cognitive effects of nicotine vapor exposure and identify cholinergic and dopaminergic mechanisms that may affected with repeated use.


Asunto(s)
Conducta de Elección/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Nicotina/toxicidad , Corteza Prefrontal/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Masculino , Agonistas Nicotínicos/toxicidad , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Receptores Nicotínicos/genética
7.
Toxicology ; 459: 152847, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34245815

RESUMEN

Previous findings have confirmed that prenatal nicotine exposure (PNE) leads to retarded cartilage development in the fetal growth plate. It is characterized by insufficient matrix synthesis and decreased expression of matrix phenotype genes aggrecan (ACAN) and Col2A1 in the fetal growth plate chondrocytes; however, the specific molecular mechanism is yet unclear. This study intends to clarify the specific molecular mechanism of fetal osteochondral retardation caused by PNE through animal and cellular experiments. The present study demonstrated that in male offspring of the PNE group (the pregnant rats were subcutaneously administered nicotine 1.0 mg/kg twice per day (2.0 mg/kg.d) at GD11-20), the cartilage matrix of the fetal growth plate was lightly stained, the collagen was reduced, and expression of the matrix phenotype genes, ACAN and Col2A1, was significantly decreased. It was further found that PNE decreased histone acetylation (H3K9/H3K14) levels in the ACAN and Col2A1 promoter regions. Moreover, the expression of Snail and HDAC1/2 was increased in the PNE group. in vitro, the nicotine treatment at different concentrations elevated the expression of Snail/HDAC1/2 while decreasing the H3K9/H3K14 levels in the ACAN and Col2A1 promoter regions. Snail-siRNA transfection partially abolished the nicotine-induced increase in HDAC1/2 expression and decreased the histone acetylation levels in the ACAN and Col2A1 promoter regions. Trichostatin A (TSA) treatment partially reversed the nicotine-induced changes in downstream parameters. In summary, PNE-induced decreased cartilage matrix synthesis in the fetal growth plate of male offspring is effectuated by Snail/HDAC1/2-mediated decreased H3K9/H3K14 levels in the ACAN and Col2A1 promoter regions.


Asunto(s)
Retardo del Crecimiento Fetal/inducido químicamente , Histona Desacetilasa 1/efectos de los fármacos , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/efectos de los fármacos , Histona Desacetilasa 2/metabolismo , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Factores de Transcripción de la Familia Snail/efectos de los fármacos , Factores de Transcripción de la Familia Snail/metabolismo , Agrecanos/metabolismo , Animales , Cartílago/efectos de los fármacos , Cartílago/patología , Condrocitos/efectos de los fármacos , Condrocitos/patología , Colágeno/metabolismo , Colágeno Tipo II/metabolismo , Femenino , Placa de Crecimiento/efectos de los fármacos , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Wistar , Transfección
8.
FASEB J ; 35(7): e21702, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34153130

RESUMEN

Perinatal smoke/nicotine exposure alters lung development and causes asthma in exposed offspring, transmitted transgenerationally. The mechanism underlying the transgenerational inheritance of perinatal smoke/nicotine-induced asthma remains unknown, but germline epigenetic modulations may play a role. Using a well-established rat model of perinatal nicotine-induced asthma, we determined the DNA methylation pattern of spermatozoa of F1 rats exposed perinatally to nicotine in F0 gestation. To identify differentially methylated regions (DMRs), reduced representation bisulfite sequencing was performed on spermatozoa of F1 litters. The top regulated gene body and promoter DMRs were tested for lung gene expression levels, and key proteins involved in lung development and repair were determined. The overall CpG methylation in F1 sperms across gene bodies, promoters, 5'-UTRs, exons, introns, and 3'-UTRs was not affected by nicotine exposure. However, the methylation levels were different between the different genomic regions. Eighty one CpG sites, 16 gene bodies, and 3 promoter regions were differentially methylated. Gene enrichment analysis of DMRs revealed pathways involved in oxidative stress, nicotine response, alveolar and brain development, and cellular signaling. Among the DMRs, Dio1 and Nmu were the most hypermethylated and hypomethylated genes, respectively. Gene expression analysis showed that the mRNA expression and DNA methylation were incongruous. Key proteins involved in lung development and repair were significantly different (FDR < 0.05) between the nicotine and placebo-treated groups. Our data show that DNA methylation is remodeled in offspring spermatozoa upon perinatal nicotine exposure. These epigenetic alterations may play a role in transgenerational inheritance of perinatal smoke/nicotine induced asthma.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Pulmón/patología , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Efectos Tardíos de la Exposición Prenatal/patología , Espermatozoides/metabolismo , Animales , Animales Recién Nacidos , Femenino , Regulación del Desarrollo de la Expresión Génica , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Ratas Sprague-Dawley , Espermatozoides/patología
9.
Toxicol Lett ; 349: 84-91, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34153408

RESUMEN

AIM: Smoking has been considered as a risk factor of chronic pancreatitis (CP), but the potential mechanism is still unknown. The major pathological feature of CP is pancreatic fibrosis, whose major functional cells are pancreatic stellate cells (PSCs). Nicotine is the major component of cigarette smoke, our recent study suggested that nicotine has the potential to facilitate pancreatic fibrosis in CP. This study was aimed to analyze the function and mechanism of nicotine on PSCs and pancreatic fibrosis in rats. MATERIALS AND METHODS: In vivo, a rat CP model was induced by intraperitoneal injection of 20 % L-arginine hydrochloride (200 mg/100 g) at 1 h intervals twice per week, nicotine was injected subcutaneously at a dose of 1 mg/kg body weight per day. After four weeks, the pancreatic tissue was collected for H&E, Masson and immunohistochemical staining. In vitro, primary rPSCs were isolated from rats and treated with nicotine (0.1 µM and 1 µM). The proliferation、apoptosis、α-SMA expression、extracellular matrix (ECM) metabolism and α7nAChR-mediated JAK2/STAT3 signaling pathway of rPSCs were detected by CCK-8 assay、flow cytometry、real-time Q-PCR and western blotting analysis. The α7nAChR antagonist α-bungarotoxin (α-BTX) was used to perform inhibition experiments. KEY FINDINGS: Nicotine increased pancreatic damage, collagen deposition and activation of PSCs in the CP rat model. In rPSCs, the proliferation, α-SMA expression and ECM formation were significantly promoted by nicotine in a dose-dependent manner. Meanwhile, the apoptosis of rPSCs was significantly reduced after nicotine treatment. Moreover, nicotine also activated the α7nAChR-mediated JAK2/STAT3 signaling pathway in rPSCs. These effects of nicotine on rPSCs were blocked by α-BTX. SIGNIFICANCE: Our finding in this research suggests that nicotine facilitates pancreatic fibrosis by promoting activation of pancreatic stellate cells via α7nAChR-mediated JAK2/STAT3 signaling pathway in rats, partly revealing the mechanism of smoking on chronic pancreatitis.


Asunto(s)
Janus Quinasa 2/metabolismo , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Células Estrelladas Pancreáticas/efectos de los fármacos , Pancreatitis Crónica/inducido químicamente , Factor de Transcripción STAT3/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibrosis , Masculino , Células Estrelladas Pancreáticas/enzimología , Células Estrelladas Pancreáticas/patología , Pancreatitis Crónica/enzimología , Pancreatitis Crónica/patología , Ratas Wistar , Transducción de Señal , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
10.
Toxicol Lett ; 349: 155-164, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34171359

RESUMEN

Cytochrome P450 1A1 (CYP1A1) is a member of a subfamily of enzymes involved in the metabolism of both endogenous and exogenous substrates and the chemical activation of xenobiotics to carcinogenic derivatives. Here, the effects of nicotine, a major psychoactive compound present in cigarette smoke, on CYP1A1 expression and human hepatocellular carcinoma (HepG2) cell proliferation were investigated. Nicotine stimulated CYP1A1 expression via the transcription factors, activator protein 1, nuclear factor-kappa B, and the aryl hydrocarbon receptor (AhR) signaling pathway. Pharmacological inhibition and mutagenesis studies indicated that p38 mitogen-activated protein kinase, as well as RelA (or p65), mediated the upregulation of CYP1A1 of nicotine in HepG2 cells. The antioxidant compound, N-acetyl-cysteine, abrogated nicotine-activated production of reactive oxygen species and inhibited CYP1A1 expression by nicotine. Furthermore, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity was inhibited by diphenyleneiodonium (an NADPH oxidase inhibitor). Thus, these results demonstrated that AhR played an important role in nicotine-induced CYP1A1 expression. Additionally, liver hepatocellular carcinoma HepG2 cells treated with nicotine exhibited markedly enhanced proliferation via CYP1A1 expression and Akt activation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma Hepatocelular/enzimología , Citocromo P-450 CYP1A1/biosíntesis , Neoplasias Hepáticas/enzimología , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Citocromo P-450 CYP1A1/genética , Inducción Enzimática , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Hidrocarburo de Aril/genética , Transducción de Señal , Factor de Transcripción AP-1/genética , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Toxicol Lett ; 343: 44-55, 2021 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-33640489

RESUMEN

Paternal nicotine exposure can alter phenotypes in future generations. The aim of this study is to explore whether paternal nicotine exposure affects the hepatic repair to chronic injury which leads to hepatic fibrosis in offspring. Our results demonstrate that nicotine down regulates mmu-miR-15b expression via the hyper-methylation on its CpG island shore region in the spermatozoa. This epigenetic modification imprinted in the liver of the offspring. The decreased mmu-miR-15b promotes the expression of Wnt4 and activates the Wnt pathway in the offspring mice liver. The activation of the Wnt pathway improves the activation and proliferation of hepatic stellate cells (HSCs) leading to liver fibrosis. Moreover, the Wnt pathway promotes the activation of the TGF-ß pathway and the two pathways cooperate to promote the transcription of extracellular matrix (ECM) genes. In conclusion, this study found that nicotine promotes hepatic fibrosis in the offspring via the activation of Wnt pathway by imprinting the hyper-methylation of mmu-miR-15b.


Asunto(s)
Cirrosis Hepática/inducido químicamente , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Exposición Paterna , Animales , Regulación hacia Abajo , Epigénesis Genética , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína Wnt4/genética , Proteína Wnt4/metabolismo
12.
Drug Test Anal ; 13(2): 242-260, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33450135

RESUMEN

Electronic cigarettes (ECs) are thought to be less harmful than traditional combustible cigarettes and were originally intended to help smokers quit. Over the past two decades, they have especially gained popularity with the younger generation. To date, there are over 7000 unique e-liquid flavours available and over 400 different e-cigarette brands. The accuracy of nicotine strength labelling in e-liquids was assessed in this work. Twenty-three studies from around the world were chosen to assess the level and frequency of nicotine mislabelling in 545 e-liquid products. Nicotine strengths were most commonly mislabelled by between 5% and 20%, with the majority testing lower than what the label indicated. Fifteen European e-liquids that were assessed were labelled as 20 mg/ml or less, yet when tested, they contained more than 20 mg/ml of nicotine. One e-liquid that was supposed to contain no nicotine in fact contained 23.91 mg/ml of nicotine. Furthermore, the difference between the medians of the available labelled and experimental nicotine concentrations was significant (p < 0.001, Wilcoxon signed rank test). Preliminary studies show that high nicotine levels delivered via aerosol increase the risk for nicotine poisoning and cause airway inflammation. Other EC ingredients, such as flavourings, contribute to EVALI and 'popcorn lung'. There is evidence that certain flavourings, such as menthol, reinforce the effects of nicotine and modify drug absorption and metabolism. There is a global need for better quality control in EC products in order to make these safe for consumers.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Etiquetado de Medicamentos , Humanos , Inflamación/inducido químicamente , Nicotina/administración & dosificación , Nicotina/efectos adversos , Nicotina/toxicidad , Agonistas Nicotínicos/administración & dosificación , Agonistas Nicotínicos/efectos adversos , Agonistas Nicotínicos/toxicidad
13.
Tuberculosis (Edinb) ; 127: 102026, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33262029

RESUMEN

Several epidemiological studies have identified the cigarette smoke as a risk factor for the infection and development of tuberculosis. Nicotine is considered the main immunomodulatory molecule of the cigarette. In the present study, we evaluated the effect of nicotine in the growth of M. tuberculosis. Lung epithelial cells and macrophages were infected with M. tuberculosis and/or treated with nicotine. The results show that nicotine increased the growth of M. tuberculosis mainly in type II pneumocytes (T2P) but not in airway basal epithelial cells nor macrophages. Further, it was observed that nicotine decreased the production of ß-defensin-2, ß-defensin-3, and the cathelicidin LL-37 in all the evaluated cells at 24 and 72 h post-infection. The modulation of the expression of antimicrobial peptides appears to be partially mediated by the nicotinic acetylcholine receptor α7 since the blockade of this receptor partially reverted the production of antimicrobial peptides. In summary, it was found that nicotine decreases the production of HBD-2, HBD-3, and LL-37 in T2P during the infection with M. tuberculosis promoting its intracellular growth.


Asunto(s)
Células Epiteliales Alveolares/microbiología , Mycobacterium tuberculosis/efectos de los fármacos , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Tuberculosis Pulmonar/microbiología , Células A549 , Células Epiteliales Alveolares/metabolismo , Péptidos Catiónicos Antimicrobianos/metabolismo , Carga Bacteriana , Interacciones Huésped-Patógeno , Humanos , Macrófagos/microbiología , Mycobacterium tuberculosis/crecimiento & desarrollo , Tuberculosis Pulmonar/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , beta-Defensinas/metabolismo , Catelicidinas
14.
Am J Physiol Heart Circ Physiol ; 320(1): H133-H143, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33216635

RESUMEN

The usage of flavored electronic nicotine delivery systems (ENDS) is popular, specifically in the teen and young adult age-groups. The possible cardiac toxicity of the flavoring aspect of ENDS is largely unknown. Vaping, a form of electronic nicotine delivery, uses "e-liquid" to generate "e-vapor," an aerosolized mixture of nicotine and/or flavors. We report our investigation into the cardiotoxic effects of flavored e-liquids. E-vapors containing flavoring aldehydes such as vanillin and cinnamaldehyde, as indicated by mass spectrometry, were more toxic in HL-1 cardiomyocytes than fruit-flavored e-vapor. Exposure of human induced pluripotent stem cell-derived cardiomyocytes to cinnamaldehyde or vanillin-flavored e-vapor affected the beating frequency and prolonged the field potential duration of these cells more than fruit-flavored e-vapor. In addition, vanillin aldehyde-flavored e-vapor reduced the human ether-à-go-go-related gene (hERG)-encoded potassium current in transfected human embryonic kidney cells. In mice, inhalation exposure to vanillin aldehyde-flavored e-vapor for 10 wk caused increased sympathetic predominance in heart rate variability measurements. In vivo inducible ventricular tachycardia was significantly longer, and in optical mapping, the magnitude of ventricular action potential duration alternans was significantly larger in the vanillin aldehyde-flavored e-vapor-exposed mice than in controls. We conclude that the widely popular flavored ENDS are not harm free, and they have a potential for cardiac harm. More studies are needed to further assess their cardiac safety profile and long-term health effects.NEW & NOTEWORTHY The use of electronic nicotine delivery systems (ENDS) is not harm free. It is not known whether ENDS negatively affect cardiac electrophysiological function. Our study in cell lines and in mice shows that ENDS can compromise cardiac electrophysiology, leading to action potential instability and inducible ventricular arrhythmias. Further investigations are necessary to assess the long-term cardiac safety profile of ENDS products in humans and to better understand how individual components of ENDS affect cardiac toxicity.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Aromatizantes/toxicidad , Frecuencia Cardíaca/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Taquicardia Ventricular/inducido químicamente , Vapeo/efectos adversos , Potenciales de Acción/efectos de los fármacos , Administración por Inhalación , Animales , Cardiotoxicidad , Canal de Potasio ERG1/metabolismo , Femenino , Aromatizantes/administración & dosificación , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatología , Factores de Tiempo
15.
Basic Clin Pharmacol Toxicol ; 128(2): 322-333, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32991780

RESUMEN

This study investigated the impact of prolonged nicotine administration on myocardial susceptibility to ischaemia-reperfusion (I/R) injury in a rat model and determined whether nicotine affects mitochondrial reactive oxygen species (ROS) production and permeability transition in rat hearts. Sprague-Dawley rats were administered 0.6 or 1.2 mg/kg nicotine for 28 days, and their hearts were isolated at end-point for assessment of myocardial susceptibility to I/R injury ex vivo. Rat heart mitochondria were also isolated from a subset of rats for analysis of mitochondrial ROS production and permeability transition. Compared to the vehicle controls, rat hearts isolated from nicotine-administered rats exhibited poorer left ventricular function that worsened over the course of I/R. Coronary flow rate was also severely impaired in the nicotine groups at baseline and this worsened after I/R. Nicotine administration significantly increased mitochondrial ROS production and permeability transition relative to the vehicle controls. Interestingly, pre-incubation of isolated mitochondria with ROS scavengers (superoxide dismutase and mitoTEMPO) significantly abolished nicotine-induced increase in mitochondria permeability transition in isolated rat heart mitochondria. Overall, our data showed that prolonged nicotine administration enhances myocardial susceptibility to I/R injury in rats and this is associated with mitochondrial ROS-driven increase in mitochondrial permeability transition.


Asunto(s)
Daño por Reperfusión Miocárdica/inducido químicamente , Miocitos Cardíacos/efectos de los fármacos , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Animales , Circulación Coronaria/efectos de los fármacos , Modelos Animales de Enfermedad , Esquema de Medicación , Preparación de Corazón Aislado , Masculino , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Función Ventricular Izquierda/efectos de los fármacos
16.
J Biol Chem ; 295(52): 18051-18064, 2020 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-33082140

RESUMEN

Evolving evidence suggests that nicotine may contribute to impaired asthma control by stimulating expression of nerve growth factor (NGF), a neurotrophin associated with airway remodeling and airway hyperresponsiveness. We explored the hypothesis that nicotine increases NGF by reducing lung fibroblast (LF) microRNA-98 (miR-98) and PPARγ levels, thus promoting airway remodeling. Levels of NGF, miR-98, PPARγ, fibronectin 1 (FN1), endothelin-1 (EDN1, herein referred to as ET-1), and collagen (COL1A1 and COL3A1) were measured in human LFs isolated from smoking donors, in mouse primary LFs exposed to nicotine (50 µg/ml), and in whole lung homogenates from mice chronically exposed to nicotine (100 µg/ml) in the drinking water. In selected studies, these pathways were manipulated in LFs with miR-98 inhibitor (anti-miR-98), miR-98 overexpression (miR-98 mimic), or the PPARγ agonist rosiglitazone. Compared with unexposed controls, nicotine increased NGF, FN1, ET-1, COL1A1, and COL3A1 expression in human and mouse LFs and mouse lung homogenates. In contrast, nicotine reduced miR-98 levels in LFs in vitro and in lung homogenates in vivo Treatment with anti-miR-98 alone was sufficient to recapitulate increases in NGF, FN1, and ET-1, whereas treatment with a miR-98 mimic significantly suppressed luciferase expression in cells transfected with a luciferase reporter linked to the putative seed sequence in the NGF 3'UTR and also abrogated nicotine-induced increases in NGF, FN1, and ET-1 in LFs. Similarly, rosiglitazone increased miR-98 and reversed nicotine-induced increases in NGF, FN1, and ET-1. Taken together, these findings demonstrate that nicotine-induced increases in NGF and other markers of airway remodeling are negatively regulated by miR-98.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/genética , Factor de Crecimiento Nervioso/metabolismo , Nicotina/toxicidad , Hipersensibilidad Respiratoria/patología , Animales , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Nervioso/genética , Agonistas Nicotínicos/toxicidad , PPAR gamma , Hipersensibilidad Respiratoria/inducido químicamente , Hipersensibilidad Respiratoria/metabolismo
17.
Am J Physiol Heart Circ Physiol ; 319(3): H651-H660, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32795172

RESUMEN

Although increased predisposition to cardiac fibrosis and cardiac dysfunction has been demonstrated in the perinatally nicotine-exposed heart, the underlying mechanisms remain unclear. With the use of a well-established rat model and cultured primary neonatal rat cardiac fibroblasts, the effect of perinatal nicotine exposure on offspring heart extracellular matrix deposition and the likely underlying mechanisms were investigated. Perinatal nicotine exposure resulted in increased collagen type I (COL1A1) and III (COL3A1) deposition along with a decrease in miR-29 family and an increase in long noncoding RNA myocardial infarction-associated transcript (MIAT) levels in offspring heart. Nicotine treatment of isolated primary neonatal rat cardiac fibroblasts suggested that these effects were mediated via nicotinic acetylcholine receptors including α7 and the induced collagens accumulation was reversed by a gain-of function of miR-29 family. Knockdown of MIAT resulted in increased miR-29 family and decreased COL1A1 and COL3A1 levels, suggesting nicotine-mediated MIAT induction as the underlying mechanism for nicotine-induced collagen deposition. Luciferase reporter assay and RNA immunoprecipitation studies showed an intense physical interaction between MIAT, miR-29 family, and argonaute 2, corroborating the mechanistic link between perinatal nicotine exposure and increased extracellular matrix deposition. Overall, perinatal nicotine exposure resulted in lower miR-29 family levels in offspring heart, while it elevated cardiac MIAT and collagen type I and III levels. These findings provide mechanistic basis for cardiac dysfunction in perinatal nicotine-exposed offspring and offer multiple novel potential therapeutic targets.NEW & NOTEWORTHY Using an established rat model and cultured primary neonatal cardiac fibroblasts, we show that nicotine mediated MIAT induction as the underlying mechanism for the excessive cardiac collagen deposition. These observations provide mechanistic basis for the increased predisposition to cardiac dysfunction following perinatal cigarette/nicotine exposure and offer novel potential therapeutic targets.


Asunto(s)
Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Cardiopatías/inducido químicamente , Miocardio/metabolismo , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Efectos Tardíos de la Exposición Prenatal , Factores de Edad , Animales , Animales Recién Nacidos , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Matriz Extracelular/patología , Femenino , Fibroblastos/patología , Fibrosis , Edad Gestacional , Cardiopatías/genética , Cardiopatías/metabolismo , Cardiopatías/patología , MicroARNs/genética , MicroARNs/metabolismo , Miocardio/patología , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Embarazo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Ratas Sprague-Dawley
18.
FASEB J ; 34(9): 11444-11459, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32654256

RESUMEN

In a rat model, perinatal nicotine exposure results in an epigenetically driven multi- and trans-generationally transmitted asthmatic phenotype that tends to wane over successive generations. However, the effect of repeat nicotine exposure during the F1 (Filial 1) gestational period on the transmitted phenotype is unknown. Using a well-established rat model, we compared lung function, mesenchymal markers of airway reactivity, and global gonadal DNA methylation changes in F2 offspring in a sex-specific manner following perinatal exposure to nicotine in only the F0 gestation, in both F0 and F1 (F0/F1) gestations, and in neither (control group). Both F0 only and F0/F1 exposure groups showed an asthmatic phenotype, an effect that was more pronounced in the F0/F1 exposure group, especially in males. Testicular global DNA methylation increased, while ovarian global DNA methylation decreased in the F0/F1 exposed group. Since the offspring of smokers are more likely to smoke than the offspring of nonsmokers, this sets the stage for more severe asthma if both mother and grandmother had smoked during their pregnancies. Increased gonadal DNA methylation changes following nicotine reexposure in the F1 generation suggests that epigenetic mechanisms might well underlie the transgenerational inheritance of acquired phenotypic traits in general and nicotine-induced asthma in particular.


Asunto(s)
Asma/diagnóstico , Pulmón/efectos de los fármacos , Nicotina/toxicidad , Efectos Tardíos de la Exposición Prenatal/diagnóstico , Animales , Asma/inducido químicamente , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Femenino , Pulmón/fisiopatología , Masculino , Exposición Materna/efectos adversos , Agonistas Nicotínicos/toxicidad , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ratas Sprague-Dawley , Pruebas de Función Respiratoria , Factores Sexuales , Testículo/efectos de los fármacos , Testículo/metabolismo
19.
Biomed Pharmacother ; 129: 110387, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32540646

RESUMEN

Approximately fifty percent of premenopausal women who smoke cigarettes or on nicotine replacement therapy are also on hormonal contraceptives, especially oral estrogen-progestin. Oral estrogen-progestin therapy has been reported to promote insulin resistance (IR) which causes lipid influx into non-adipose tissue and impairs Na+/K+ -ATPase activity, especially in the heart and kidney. However, the effects of nicotine on excess lipid and altered Na+/K+ -ATPase activity associated with the use of estrogen-progestin therapy have not been fully elucidated. This study therefore aimed at investigating the effect of nicotine on cardiac and renal lipid influx and Na+/K+ -ATPase activity during estrogen-progestin therapy. Twenty-four female Wistar rats grouped into 4 (n = 6/group) received (p.o.) vehicle, nicotine (1.0 mg/kg) with or without estrogen-progestin steroids (1.0 µg ethinyl estradiol and 5.0 µg levonorgestrel) and estrogen-progestin only daily for 6 weeks. Data showed that estrogen-progestin treatment or nicotine exposure caused IR, hyperinsulinemia, increased cardiac and renal uric acid, malondialdehyde, triglyceride, glycogen synthase kinase-3, plasminogen activator inhibitor-1, reduced bilirubin and circulating estradiol. Estrogen-progestin treatment led to decreased cardiac Na+/K+-ATPase activity while nicotine did not alter Na+/K+-ATPase activity but increased plasma and tissue cotinine. Renal Na+/K+-ATPase activity was not altered by the treatments. However, all these alterations were reversed following combined administration of oral estrogen-progestin therapy and nicotine. The present study therefore demonstrates that oral estrogen-progestin therapy and nicotine exposure synergistically prevents IR-linked cardio-renotoxicity with corresponding improvement in cardiac and renal lipid accumulation, oxidative stress, inflammation and Na+/K+-ATPase activity.


Asunto(s)
Anticonceptivos Orales Combinados/farmacología , Estrógenos/farmacología , Etinilestradiol/farmacología , Corazón/efectos de los fármacos , Riñón/efectos de los fármacos , Levonorgestrel/farmacología , Miocardio/enzimología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Progestinas/farmacología , Animales , Anticonceptivos Orales Combinados/toxicidad , Citoprotección , Combinación de Medicamentos , Sinergismo Farmacológico , Estrógenos/toxicidad , Etinilestradiol/toxicidad , Femenino , Mediadores de Inflamación/metabolismo , Riñón/enzimología , Riñón/patología , Levonorgestrel/toxicidad , Metabolismo de los Lípidos/efectos de los fármacos , Miocardio/patología , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Estrés Oxidativo/efectos de los fármacos , Progestinas/toxicidad , Ratas Wistar , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
20.
Reprod Biol Endocrinol ; 18(1): 65, 2020 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-32552695

RESUMEN

BACKGROUND: Nicotine, a pharmacologically active component of tobacco adversely affects the male reproductive system and fertility whereas icariin (ICA), the main active ingredient in Epimedium herba has been used in the treatment of several male reproductive problems. This study aimed at evaluating the protective or ameliorative effect of ICA against reproductive toxicity induced by intraperitoneal injection of nicotine in mice. METHODS: Using simple random allocation, forty male mice were randomly divided into 4 groups: control (received 0.35 mL physiological saline via gastric gavage), nicotine (0.75 mg/kg BW/day intraperitoneally), ICA (75 mg/kg BW/day gastric gavage), and nicotine plus ICA (nicotine, 0.75 mg/kg BW/day intraperitoneally + ICA, 75 mg/kg BW/day gastric gavage) group. After 35 days of treatment, the mice were weighed, sacrificed, and their reproductive organs (testis and epididymis) were collected and examined for further studies. RESULTS: The nicotine-treated group showed significantly decreased epididymal sperm density and serum testosterone concentration relative to the control group. Nicotine also caused oxidative damage shown by significant reduction in the activities of antioxidant enzymes and elevation in Malondialdehyde (MDA) levels. ICA on the other hand, improved the reduction in sperm density, hormone levels, and activities of antioxidant enzymes altered in the nicotine treated mice. CONCLUSIONS: These findings indicate that nicotine-induced reproductive toxicity and oxidative damage on male reproductive tissues could be attenuated by ICA.


Asunto(s)
Epidídimo/efectos de los fármacos , Flavonoides/farmacología , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Sustancias Protectoras/farmacología , Espermatozoides/efectos de los fármacos , Testículo/efectos de los fármacos , Testosterona/sangre , 3-Hidroxiesteroide Deshidrogenasas/efectos de los fármacos , 3-Hidroxiesteroide Deshidrogenasas/genética , Animales , Peso Corporal/efectos de los fármacos , Epidídimo/patología , Glutatión Peroxidasa/efectos de los fármacos , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Tamaño de los Órganos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fosfoproteínas/efectos de los fármacos , Fosfoproteínas/genética , Distribución Aleatoria , Recuento de Espermatozoides , Espermatozoides/patología , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Testículo/metabolismo , Testículo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...