Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 262
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Toxicol Appl Pharmacol ; 486: 116937, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38643950

RESUMO

Selective Serotonin Reuptake Inhibitors (SSRIs) are widely used medications for the treatment of major depressive disorder. However, long-term SSRI use has been associated with weight gain and altered lipid profiles. These findings suggest that SSRIs may have negative effects on metabolism. Exposure to certain chemicals called 'obesogens' is known to promote lipid accumulation and obesity by modulating adipogenesis. Here, we investigated whether citalopram (CIT) and sertraline (SER) interfere with the process of adipogenesis, using human mesenchymal stem cells (MSCs) in a 2D and a 3D model. Assessment of intracellular lipid accumulation by fluorescence staining was used as a measure for enhanced adipogenesis. To explore possible mechanisms behind SSRIs' effects, receptor mediated activity was studied using responsive cell lines for various nuclear receptors. Furthermore, RNA sequencing was performed in the 3D model, followed by differential gene expression and pathway analysis. A dose dependent increase in lipid accumulation was observed in both models with CIT and SER. For the 3D model, the effect was seen in a range close to reported steady-state plasma concentrations (0.065-0.65 µM for SER and 0.12-0.92 µM for CIT). Pathway analysis revealed unexpected results of downregulation in adipogenesis-related pathways and upregulation in phospholipids and lysosomal pathways. This was confirmed by an observed increase in lysosomes in the 2D model. Our findings suggest lysosomal dysfunction and disrupted lipid metabolism in mature adipocytes, leading to excessive phospholipid synthesis. Moreover, important adipogenic processes are inhibited, potentially leading to dysfunctional adipocytes, which might have implications in the maintenance of a healthy metabolic balance.


Assuntos
Adipogenia , Antidepressivos , Citalopram , Metabolismo dos Lipídeos , Células-Tronco Mesenquimais , Inibidores Seletivos de Recaptação de Serotonina , Sertralina , Adipogenia/efeitos dos fármacos , Sertralina/farmacologia , Sertralina/toxicidade , Humanos , Citalopram/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Antidepressivos/farmacologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga
2.
Am J Physiol Gastrointest Liver Physiol ; 325(6): G528-G538, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37724979

RESUMO

Perinatal exposure to selective serotonin reuptake inhibitors (SSRIs) has been shown to disrupt the development of serotonergic signaling pathways in the brain and enteric nervous system. Serotonin (5-hydroxytryptamine; 5-HT) signaling is critical for gastrointestinal homeostasis; changes in 5-HT expression and regulation have been associated with gastrointestinal diseases of motility and inflammation. We tested the hypothesis that perinatal exposure to the SSRI fluoxetine can influence the development of the gastrointestinal tract in exposed offspring. Female nulliparous Wistar rats were given fluoxetine (10 mg/kg) or vehicle control from 2 wk before mating until weaning; small and large intestines of female and male offspring were collected at postnatal days 1, 21 (P1, P21, respectively), and 6 mo of age. In histological preparations, the proportion of serotonergic neurons significantly increased in the colons of both female and male fluoxetine-exposed compared with control offspring at P21, a time point that signifies maximal exposure to fluoxetine. At 6 mo of age, male but not female fluoxetine-exposed offspring had a significant increase in circulating 5-HT, with a significant decrease in transcripts encoding the 5-HT2A receptor and monoamine oxidase as compared with control offspring. Measurement of spatiotemporal mapping of contractile activity of the small and large intestine at 6 mo of age revealed no changes in motility in the small bowel of fluoxetine-exposed offspring but revealed a significant increase in the frequency of colonic contractions in the female fluoxetine-exposed compared with control animals. Susceptibility to inflammation was examined at 6 mo using the dextran sulfate sodium model of acute colitis. In utero exposure to fluoxetine was not found to exacerbate colitis severity. These findings suggest that fluoxetine exposure during fetal and early postnatal development can lead to changes in serotonergic neurons at the peak of exposure with sex-specific changes in 5-HT signaling and colonic motility in adulthood.NEW & NOTEWORTHY There is increasing recognition of the relevance of in utero and early postnatal exposures in the developmental programming of the gastrointestinal tract. Perinatal exposure to selective serotonin reuptake inhibitors and antidepressant medications is of particular relevance as they are commonly prescribed during pregnancy, and serotonergic pathways play key roles during gastrointestinal development and in postnatal homeostasis. Here, we provide a comprehensive evaluation of clinically relevant outcomes of gastrointestinal motility and susceptibility to colitis in fluoxetine-exposed offspring and highlight changes in colonic serotonergic neurons at the peak of perinatal fluoxetine exposure with sex-dependent changes in serotonin signaling and colonic motility in adulthood.


Assuntos
Colite , Efeitos Tardios da Exposição Pré-Natal , Gravidez , Humanos , Ratos , Animais , Masculino , Feminino , Fluoxetina/toxicidade , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Serotonina/metabolismo , Ratos Sprague-Dawley , Ratos Wistar , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Inflamação , Colite/induzido quimicamente
3.
Ecotoxicol Environ Saf ; 250: 114493, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36608562

RESUMO

Antidepressant prescriptions are on a rise worldwide and this increases the concerns for the impacts of these pharmaceuticals on nontarget organisms. Antidepressants are neuroactive compounds that can affect organism's behavior. Behavior is a sensitive endpoint that may also propagate effects at a population level. Another interesting aspect of antidepressants is that they have shown to induce non-monotonic dose-response (NMDR) curves. While such NMDR relationships may have clear implications for the environmental risk, the resolution of current studies is often too coarse to be able to detect relevant NMDR. Therefore, the current study was performed into the behavioral effects (activity, feeding and chemotaxis) in Caenorhabditis elegans as the model organism of the selective serotonin reuptake inhibitors fluoxetine and sertraline and the acetylcholinesterase inhibiting pesticide chlorpyrifos, using a wide range of concentrations (ng/l to mg/l). In order to statistically examine the non-monotonicity, nonlinear regression models were applied to the results. The results showed a triphasic dose-response relationship for activity and chemotaxis after exposure to fluoxetine, but not to sertraline or chlorpyrifos. Effects of fluoxetine already occurred at low concentrations in the range of ng/l while sertraline only showed effects at concentrations in the µg/l range, similar to chlorpyrifos. The different responses between fluoxetine and sertraline, both SSRIs, indicate that response patterns may not always be extrapolated from chemicals with the same primary mode of action. The effects of fluoxetine at low concentrations, in a non-monotonic manner, confirm the relevance of examining such responses at low concentrations.


Assuntos
Clorpirifos , Fluoxetina , Animais , Fluoxetina/toxicidade , Sertralina/toxicidade , Caenorhabditis elegans , Acetilcolinesterase , Antidepressivos/toxicidade , Inibidores Seletivos de Recaptação de Serotonina/toxicidade
4.
J Cardiovasc Pharmacol ; 80(2): 261-269, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35904815

RESUMO

ABSTRACT: Selective serotonin reuptake inhibitors (SSRIs) are antidepressants prescribed in 10% of pregnancies in the United States. Maternal use of SSRIs has been linked to an elevated rate of congenital heart defects, but the exact mechanism of pathogenesis is unknown. Previously, we have shown a decrease in cardiomyocyte proliferation, left ventricle size, and reduced cardiac expression of the serotonin receptor 5-HT 2B in offspring of mice exposed to the SSRI sertraline during pregnancy, relative to offspring of untreated mice. These results suggest that disruption of serotonin signaling leads to heart defects. Supporting this conclusion, we show here that zebrafish embryos exposed to sertraline develop with a smaller ventricle, reduced cardiomyocyte number, and lower cardiac expression of htr2b relative to untreated embryos. Moreover, zebrafish embryos homozygous for a nonsense mutation of htr2b ( htr2bsa16649 ) were sensitized to sertraline treatment relative to wild-type embryos. Specifically, the ventricle area was reduced in the homozygous htr2b mutants treated with sertraline compared with wild-type embryos treated with sertraline and homozygous htr2b mutants treated with vehicle control. Whereas long-term effects on left ventricle shortening fraction and stroke volume were observed by echocardiography in adult mice exposed to sertraline in utero, echocardiograms of adult zebrafish exposed to sertraline as embryos were normal. These results implicate the 5-HT 2B receptor functions in heart development and suggest zebrafish are a relevant animal model that can be used to investigate the connection between maternal SSRI use and elevated risk of congenital heart defects.


Assuntos
Cardiopatias Congênitas , Sertralina , Animais , Feminino , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/genética , Camundongos , Miócitos Cardíacos/metabolismo , Gravidez , Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Sertralina/toxicidade , Peixe-Zebra/genética
5.
Environ Sci Technol ; 56(17): 12380-12390, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-35985052

RESUMO

Residual antidepressants are of increasing concern worldwide, yet critical information on their long-term neurotoxic impacts on nontarget aquatic animals is lacking. Here, we investigated the long-term effects (from 0 to 150 days postfertilization) of the selective serotonin reuptake inhibitor citalopram (0.1-100 µg/L) on motor function, learning, and memory in zebrafish over two generations and explored the reversibility of the effect in F1 larvae. Unlike F0+ larvae, we found that F1+ larvae displayed decreased sensorimotor performance when continuously exposed to citalopram at 100 µg/L. No adverse effects were found in F1- larvae after they were transferred to a clean medium. Whole-mount immunofluorescence assays suggested that the motor impairments were related to axonal projections of the spinal motor neurons (MNs). For F0+ adults, long-term citalopram exposure mainly caused male-specific declines in motor, learning, and memory performance. Analysis of serotonergic and cholinergic MNs revealed no significant changes in the male zebrafish spinal cord. In contrast, the number of glutamatergic spinal MNs decreased, likely associated with the impairment of motor function. Additionally, treatment with 100 µg/L citalopram significantly reduced the number of dopaminergic neurons, but no significant neuronal apoptosis was observed in the adult telencephalon. Overall, this study provides neurobehavioral evidence and novel insights into the neurotoxic mechanisms of long-term citalopram exposure and may facilitate the assessment of the environmental and health risks posed by citalopram-containing antidepressant drugs.


Assuntos
Citalopram , Inibidores Seletivos de Recaptação de Serotonina , Animais , Antidepressivos , Citalopram/toxicidade , Larva , Masculino , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Peixe-Zebra
6.
J Neuroinflammation ; 18(1): 47, 2021 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-33602262

RESUMO

BACKGROUND: Selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs) are commonly used new-generation drugs for depression. Depressive symptoms are thought to be closely related to neuroinflammation. In this study, we used up-to-date protocols of culture and stimulation and aimed to understand how astrocytes respond to the antidepressants. METHODS: Primary astrocytes were isolated and cultured using neurobasal-based serum-free medium. The cells were treated with a cytokine mixture comprising complement component 1q, tumor necrosis factor α, and interleukin 1α with or without pretreatments of antidepressants. Cell viability, phenotypes, inflammatory responses, and the underlying mechanisms were analyzed. RESULTS: All the SSRIs, including paroxetine, fluoxetine, sertraline, citalopram, and fluvoxamine, show a visible cytotoxicity within the range of applied doses, and a paradoxical effect on astrocytic inflammatory responses as manifested by the promotion of inducible nitric oxide synthase (iNOS) and/or nitric oxide (NO) and the inhibition of interleukin 6 (IL-6) and/or interleukin 1ß (IL-1ß). The SNRI venlafaxine was the least toxic to astrocytes and inhibited the production of IL-6 and IL-1ß but with no impact on iNOS and NO. All the drugs had no regulation on the polarization of astrocytic A1 and A2 types. Mechanisms associated with the antidepressants in astrocytic inflammation route via inhibition of JNK1 activation and STAT3 basal activity. CONCLUSIONS: The study demonstrated that the antidepressants possess differential cytotoxicity to astrocytes and function differently, also paradoxically for the SSRIs, to astrocytic inflammation. Our results provide novel pieces into understanding the differential efficacy and tolerability of the antidepressants in treating patients in the context of astrocytes.


Assuntos
Antidepressivos/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Animais , Animais Recém-Nascidos , Antidepressivos/toxicidade , Astrócitos/patologia , Células Cultivadas , Relação Dose-Resposta a Droga , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Ratos , Ratos Sprague-Dawley , Inibidores Seletivos de Recaptação de Serotonina/toxicidade
7.
Toxicol Appl Pharmacol ; 419: 115513, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33785354

RESUMO

The early characterization of ligands at the dopamine and serotonin transporters, DAT and SERT, respectively, is important for drug discovery, forensic sciences, and drug abuse research. 4-Methyl amphetamine (4-MA) is a good example of an abused drug whose overdose can be fatal. It is a potent substrate at DAT and SERT where its simplest secondary amine (N-methyl 4-MA) retains substrate activity at them. In contrast, N-n-butyl 4-MA is very weak, therefore it was categorized as inactive at these transporters. Here, N-octyl 4-MA and other related compounds were synthesized, and their activities were evaluated at DAT and SERT. To expedite this endeavor, cells expressing DAT or SERT were co-transfected with a voltage-gated Ca2+ channel and, the genetically-encoded Ca2+ sensor, GCaMP6s. Control compounds and the newly synthesized molecules were tested on these cells using an automated multi-well fluorescence plate reader; substrates and inhibitors were identified successfully at DAT and SERT. N-Octyl 4-MA and three bivalent compounds were inhibitors at these transporters. These findings were validated by measuring Ca2+-mobilization using quantitative fluorescence microscopy. The bivalent molecules were the most potent of the series and were further characterized in an uptake-inhibition assay. Compared to cocaine, they showed comparable potency inhibiting uptake at DAT and higher potency at SERT. These observations support a previous hypothesis that amphetamine-related (and, here, N-extended alkyl and) bivalent arylalkylamine molecules are active at monoamine transporters, showing potent activity as reuptake inhibitors, and implicate the involvement of a distant auxiliary binding feature to account for their actions at DAT and SERT.


Assuntos
Técnicas Biossensoriais , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/antagonistas & inibidores , Inibidores da Captação de Dopamina/toxicidade , Proteínas de Fluorescência Verde/metabolismo , Metanfetamina/toxicidade , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Proteínas da Membrana Plasmática de Transporte de Serotonina/efeitos dos fármacos , Canais de Cálcio/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Metanfetamina/análogos & derivados , Metanfetamina/síntese química , Microscopia de Fluorescência , Estrutura Molecular , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Relação Estrutura-Atividade , Fatores de Tempo
8.
Environ Sci Technol ; 55(11): 7479-7490, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34002605

RESUMO

The increasing number of people with depression worldwide has led to concerns regarding antidepressant contamination in aquatic environments, which could have the risk of negative effects on aquatic organisms. Chirality increases its toxicity potentials. Accordingly, we investigated the negative effects of racemic (rac-), R-, and S-FX at environmental levels (100 ng/L) on the brain serotonergic system in zebrafish (Danio rerio) for 42 days. Additionally, we measured the whole-body concentrations of FX and norfluoxetine (NFX). We found that S-FX exposure disrupted the brain serotonergic system more severely than rac- and R-FX exposure. The mechanism underlying this disruption induced by S-FX was sex-specific, with female zebrafish showing disruption of the serotonin (5-HT) release process but male zebrafish showing disruption of the 5-HT synthesis process. In addition, enantioselective enrichment and biotransformation (R-FX to R-NFX and S-FX to S-NFX) occurred in zebrafish. Sex-specific accumulation was also observed, with higher concentrations in females. Our study provides evidence for enantiomer- and sex-specific effects of FX exposure at biologically relevant concentrations. More broadly, our study demonstrated that SSRI antidepressants, such as FX, can affect aquatic life by causing important shifts in not only their active sites of the serotonin transporter.


Assuntos
Fluoxetina , Poluentes Químicos da Água , Animais , Encéfalo , Feminino , Fluoxetina/toxicidade , Masculino , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Estereoisomerismo , Poluentes Químicos da Água/toxicidade , Peixe-Zebra
9.
J Appl Toxicol ; 41(8): 1232-1240, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33179799

RESUMO

Fluoxetine is a first-line selective serotonin reuptake inhibitor widely applied for the treatment of depression; however, it induces abnormal hepatic lipid metabolism. Considering decreased expression or function of glucose-6-phosphatase (G6Pase), a key enzyme in gluconeogenesis, or the upregulation of fatty acid uptake, causes hepatic lipid accumulation. The aim of this study was to elucidate whether G6Pase regulation and fatty acid uptake alteration contribute to fluoxetine-induced abnormal hepatic lipid metabolism. Our study revealed that 8-week oral administration of fluoxetine dose-dependently increased hepatic triglyceride, causing hepatic steatosis. Concomitantly, the expression of G6Pase in mouse livers and primary mouse hepatocytes (PMHs) was downregulated in a concentration-dependent manner. Furthermore, fluoxetine increased the concentrations of glucose-6-phosphate (G6Pase substrate) and acetyl CoA (the substrate for de novo lipogenesis) in mouse livers. Additionally, fluoxetine also induced lipid accumulation and downregulated G6Pase expression in HepG2 cells. However, the uptake of green fluorescent fatty acid (BODIPY™ FL C16) in PMHs was not changed after fluoxetine treatment, indicating that fluoxetine-induced hepatic steatosis was not associated with fatty acid uptake alteration. In conclusion, fluoxetine downregulated hepatic G6Pase expression, subsequently enhanced the transformation of glucose to lipid, and ultimately resulted in hepatic steatosis, but with no impact on fatty acid uptake.


Assuntos
Fígado Gorduroso/induzido quimicamente , Fluoxetina/toxicidade , Glucose-6-Fosfatase/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Animais , Relação Dose-Resposta a Droga , Regulação para Baixo , Fígado Gorduroso/enzimologia , Fígado Gorduroso/metabolismo , Células Hep G2/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Humanos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Triglicerídeos/metabolismo
10.
Ecotoxicol Environ Saf ; 210: 111868, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33421720

RESUMO

Psychiatric drugs are among the leading medications prescribed for humans, with their presence in aquatic environments raising concerns relating to potentially harmful effects on non-target organisms. Nortriptyline (NTP) is a selective serotonin-norepinephrine reuptake inhibitor antidepressant, widely used in clinics and found in environmental water matrices. In this study, we evaluated the toxic effects of NTP on zebrafish (Danio rerio) embryos and early larval stages. Developmental and mortality analyses were performed on zebrafish exposed to NTP for 168 h at concentrations ranging from 500 to 46,900 µg/L. Locomotor behaviour and acetylcholinesterase (AChE) activity were evaluated by exposing embryos/larvae to lower NTP concentrations (0.006-500 µg/L). The median lethal NTP concentration after 168 h exposure was 2190 µg/L. Although we did not identify significant developmental changes in the treated groups, lack of equilibrium was already visible in surviving larvae exposed to ≥ 500 µg/L NTP. The behavioural analyses showed that NTP was capable of modifying zebrafish larvae swimming behaviour, even at extremely low (0.006 and 0.088 µg/L) environmentally relevant concentrations. We consistently observed a significant reduction in AChE activity in the animals exposed to 500 µg/L NTP. Our results highlight acute toxic effects of NTP on the early-life stages of zebrafish. Most importantly, exposure to environmentally relevant NTP concentrations may affect zebrafish larvae locomotor behaviour, which in turn could reduce the fitness of the species. More studies involving chronic exposure and sensitive endpoints are warranted to better understand the effect of NTP in a more realistic exposure scenario.


Assuntos
Inibidores da Captação Adrenérgica/toxicidade , Antidepressivos Tricíclicos/toxicidade , Nortriptilina/toxicidade , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Poluentes Químicos da Água/toxicidade , Peixe-Zebra , Acetilcolinesterase/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Embrião não Mamífero/efeitos dos fármacos , Larva/efeitos dos fármacos , Locomoção/efeitos dos fármacos
11.
Arch Toxicol ; 94(5): 1417-1441, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32246176

RESUMO

Depressive disorders are amongst the greatest mental health challenges, with an increasing number of patients being diagnosed each year. Though it has not yet been fully elucidated, redox metabolism imbalances and oxidative stress seem to play a major role in the pathogenesis of depressive disorders. Selective serotonin reuptake inhibitors (SSRIs) are the most prescribed antidepressants, considered to have a better tolerability. However, several adverse effects have been reported and the mechanisms involved in their pharmacological activity are not entirely understood. SSRIs have been shown to influence the redox metabolism, which could be involved in their toxicity and pharmacological effects. A comparative analysis of published in vivo and in vitro data regarding the activity of SSRIs on the redox metabolism pathways has been performed in this paper, with an emphasis on mechanistical aspects. Furthermore, a comparison between oxidative stress biomarker levels reported by different studies was attempted. The reviewed data point towards both pro- and antioxidant effects of SSRIs, dependent on tissue/cell type and dose/concentration, suggest a redox modulating potential of these compounds. In hepatic and testicular tissue, the majority of reviewed studies reported pro-oxidant effects, with possible implications towards the hepatotoxicity and sexual dysfunction that were reported following SSRI treatment; while in brain, the most common findings were antioxidant effects that could partially explain their antidepressant activity. However, given the heterogeneity of the reviewed data, further research is needed to fully understand the impact of SSRIs on redox metabolism and its implications.


Assuntos
Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Antidepressivos/farmacologia , Antidepressivos/toxicidade , Humanos , Oxirredução , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
12.
Arch Toxicol ; 94(7): 2401-2411, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32372212

RESUMO

Sertraline, an antidepressant, is commonly used to manage mental health symptoms related to depression, anxiety disorders, and obsessive-compulsive disorder. The use of sertraline has been associated with rare but severe hepatotoxicity. Previous research demonstrated that mitochondrial dysfunction, apoptosis, and endoplasmic reticulum stress were involved in sertraline-associated cytotoxicity. In this study, we reported that after a 24-h treatment in HepG2 cells, sertraline caused cytotoxicity, suppressed topoisomerase I and IIα, and damaged DNA in a concentration-dependent manner. We also investigated the role of cytochrome P450 (CYP)-mediated metabolism in sertraline-induced toxicity using our previously established HepG2 cell lines individually expressing 14 CYPs (1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5, and 3A7). We demonstrated that CYP2D6, 2C19, 2B6, and 2C9 metabolize sertraline, and sertraline-induced cytotoxicity was significantly decreased in the cells expressing these CYPs. Western blot analysis demonstrated that the induction of É£H2A.X (a hallmark of DNA damage) and topoisomerase inhibition were partially reversed in CYP2D6-, 2C19-, 2B6-, and 2C9-overexpressing HepG2 cells. These data indicate that DNA damage and topoisomerase inhibition are involved in sertraline-induced cytotoxicity and that CYPs-mediated metabolism plays a role in decreasing the toxicity of sertraline.


Assuntos
Antidepressivos/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Sistema Enzimático do Citocromo P-450/metabolismo , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Sertralina/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Dano ao DNA , DNA Topoisomerases Tipo I/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Células Hep G2 , Hepatócitos/enzimologia , Hepatócitos/patologia , Humanos , Isoenzimas , Fígado/enzimologia , Fígado/patologia , Desintoxicação Metabólica Fase I , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo
13.
Metab Brain Dis ; 35(8): 1341-1351, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32827287

RESUMO

Serotonin exerts a significant role in the mammalian central nervous system embryogenesis and brain ontogeny. Therefore, we investigate the effect of perinatal fluoxetine (FLX), a selective serotonin reuptake inhibitor, administration on the behavioral expression of adult male Swiss mice. For this purpose, two groups (n = 6 each, and ~ 35 g) of pregnant female Swiss mice were mated. Their offspring were treated with FLX (10 mg/Kg, s.c.) from postnatal day (PND) 5 to 15. At PND 16, one male puppy of each litter was euthanized, and the hippocampus was dissected for RNA analysis. At 70 days of life, the male offspring underwent a behavioral assessment in the open field, object recognition task, light-dark box, tail suspension and rotarod test. According to our results, the programmed animals had a decrease in TPH2, 5HT1a, SERT, BDNF, and LMX1B expression. Also, it was observed less time of immobility in tail suspension test and higher grooming time in the open field test. In the light-dark box test, the FLX-treated offspring had less time in the light side than control. We also observed a low cognitive performance in the object recognition task and poor motor skill learning in the rotarod test. These findings suggest that programming with FLX during the neonatal period alters a hippocampal serotonergic system, promoting anxiety and antidepressant behavior in adults, as well as a low mnemonic capacity.


Assuntos
Ansiedade/induzido quimicamente , Ansiedade/metabolismo , Fluoxetina/toxicidade , Hipocampo/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Animais , Animais Recém-Nascidos , Ansiedade/psicologia , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Feminino , Fluoxetina/administração & dosagem , Hipocampo/metabolismo , Masculino , Camundongos , Gravidez , Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Fatores de Tempo
14.
Ecotoxicol Environ Saf ; 203: 110934, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32888599

RESUMO

Pharmaceuticals and personal care products are emerging contaminants that are increasingly detected in the environment worldwide. Certain classes of pharmaceuticals, such as selective serotonin reuptake inhibitors (SSRIs), are a major environmental concern due to their widespread use and the fact that these compounds are designed to have biological effects at low doses. A complication in predicting toxic effects of SSRIs in nontarget organisms is that their mechanism of action is not fully understood. To better understand the potential toxic effects of SSRIs, we employed an ultra-low input RNA-sequencing method to identify potential pathways that are affected by early exposure to two SSRIs (fluoxetine and paroxetine). We exposed wildtype zebrafish (Danio rerio) embryos to 100 µg/L of either fluoxetine or paroxetine for 6 days before extracting and sequencing mRNA from individual larval brains. Differential gene expression analysis identified 1550 genes that were significantly affected by SSRI exposure with a core set of 138 genes altered by both SSRIs. Weighted gene co-expression network analysis identified 7 modules of genes whose expression patterns were significantly correlated with SSRI exposure. Functional enrichment analysis of differentially expressed genes as well as network module genes repeatedly identified various terms associated with mitochondrial and neuronal structures, mitochondrial respiration, and neurodevelopmental processes. The enrichment of these terms indicates that toxic effects of SSRI exposure are likely caused by mitochondrial dysfunction and subsequent neurodevelopmental effects. To our knowledge, this is the first effort to study the tissue-specific transcriptomic effects of SSRIs in developing zebrafish, providing specific, high resolution molecular data regarding the sublethal effects of SSRI exposure.


Assuntos
Encéfalo/efeitos dos fármacos , Larva/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Transcriptoma/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Peixe-Zebra , Animais , Encéfalo/embriologia , Biologia Computacional , Embrião não Mamífero/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Humanos , Larva/genética , Análise de Sequência de RNA , Peixe-Zebra/genética
15.
Arch Environ Contam Toxicol ; 79(4): 478-487, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33151376

RESUMO

Selective serotonin reuptake inhibitors (SSRIs) are psychotropic pharmaceuticals used as antidepressants. SSRIs are commonly found in surface waters in populated areas across the globe. They exert their effect by blocking the serotonin re-uptake transporter in the presynaptic nerve ending. The present study examined whether behavioural effects to exposure to SSRI citalopram depend on personality and sex in the stickleback (Gasterosteus aculeatus). Three aspects of stickleback behaviour are examined: feeding behaviour, aggression, and boldness. We exposed sticklebacks to 350-380 ng/l citalopram for 3 weeks. Feeding and aggressive behaviour were recorded before and after exposure, whereas scototaxis behaviour was tested after exposure. The results show treatment effects in feeding and aggressive behaviour. Feeding is suppressed only in the male group (χ2 = 20.4, P < 0.001) but not in the females (χ2 = 0.91, P = 0.339). Aggressive behaviour was significantly affected by treatment (χ2 = 161.9, P < 0.001), sex (χ2 = 86.3, P < 0.001), and baseline value (χ2 = 58.8, P < 0.001). Aggressiveness was suppressed by citalopram treatment. In addition, the fish showed no change in aggression and feeding behaviour over time regardless of sex and treatment, which indicate personality traits. Only females are affected by treatment in the scototaxis test. The exposed females spent significantly (χ2 = 5.02, P = 0.050) less time in the white zone than the female controls.


Assuntos
Citalopram/toxicidade , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Smegmamorpha/fisiologia , Poluentes Químicos da Água/toxicidade , Agressão/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Citalopram/farmacologia , Comportamento Alimentar , Feminino , Masculino , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Coluna Vertebral
16.
Pak J Pharm Sci ; 33(3): 1169-1172, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-33191244

RESUMO

Chemotherapy, radiotherapy, surgery and depression are the conditions that run in parallel fashions. All these conditions cause the release of an increased amount of serotonin in the body. Serotonin acts on these 5HT3 receptors and causes nausea and vomiting. Ondansetron acts by blocking serotonin from acting on the receptors and thus is useful in decreasing episodes of nausea and vomiting but when used concomitantly with SSRIs (selective serotonin reuptake inhibitors) as cancer patient also suffered from depression. This combination tends to decrease the efficacy of ondansetron. The present study was carried out to observe the modulatory role of ondansetron on ileal smooth muscle motility in vitro. Experiments were performed in four groups (n=6) and ileal smooth muscle activity was recorded on the power lab (USA). The effects of increasing concentrations of serotonin, ondansetron and paroxetine alone were observed. In the fourth group effects of paroxetine in the presence of fixed concentration (1ml) of ondansetron (10-6M) was observed. The maximum response obtained by serotonin served as a control for our study (100%). Paroxetine response on intestinal motility was completely blocked in the presence of ondansetron. Our findings hence, reinforce the hypothesis that paroxetine decreases the antiemetic activity of serotonin antagonist ondansetron, by super sensitization of serotonergic receptors resulting in an increased incidence of nausea and vomiting in cancer patient despite adequate antiemetic prophylaxis.


Assuntos
Antieméticos/farmacologia , Motilidade Gastrointestinal/efeitos dos fármacos , Íleo/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Ondansetron/farmacologia , Paroxetina/farmacologia , Receptores de Serotonina/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Antagonistas da Serotonina/farmacologia , Animais , Interações Medicamentosas , Feminino , Íleo/metabolismo , Masculino , Músculo Liso/metabolismo , Náusea/induzido quimicamente , Náusea/metabolismo , Náusea/fisiopatologia , Paroxetina/toxicidade , Coelhos , Receptores de Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Vômito/induzido quimicamente , Vômito/metabolismo , Vômito/fisiopatologia
17.
J Pharmacol Exp Ther ; 371(1): 113-120, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31308195

RESUMO

Long-term use of selective serotonin reuptake inhibitors (SSRIs) targeting the serotonin transporter (SERT) has been suggested to be associated with an increased risk for obesity and type 2 diabetes. Previously, using a murine knockout model of SERT, we showed that estrogen suppression is involved in SERT deficiency-induced obesity and glucose intolerance in nonpregnant mice. The present study investigated the effects of chronic paroxetine treatment on adiposity and glucose tolerance in mice before and during pregnancy. Chronic paroxetine treatment in nonpregnant mice resulted in visceral adiposity and glucose intolerance accompanied by reduced circulating 17ß-estradiol levels and ovarian expression of the aromatase (CYP19a1). Remarkably, pregnancy significantly reduced adiposity and improved glucose tolerance in paroxetine-treated mice by rebooting ovarian CYP19a1 expression and 17ß-estradiol production. These effects appear to be reversible as ovarian CYP19a1 expression and circulating 17ß-estradiol returned to prepregnancy levels soon after parturition. As in pregnant mice, 17ß-estradiol replacement treatment in nonpregnant mice reduced paroxetine-induced adiposity. Our findings further suggested that modulation of estrogen synthesis underlies the observed metabolic adverse effects of SSRIs. Although our data revealed a transient reversal effect of pregnancy on SSRI-induced metabolic abnormalities, these observations are experimental and limited to mice. The use of SSRIs during human pregnancy should be cautioned because of potential adverse effects to the fetuses.


Assuntos
Adiposidade , Intolerância à Glucose , Obesidade/induzido quimicamente , Paroxetina/efeitos adversos , Complicações na Gravidez/induzido quimicamente , Inibidores Seletivos de Recaptação de Serotonina/efeitos adversos , Animais , Aromatase/genética , Aromatase/metabolismo , Estradiol/metabolismo , Estradiol/uso terapêutico , Feminino , Terapia de Reposição Hormonal , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/tratamento farmacológico , Ovário/metabolismo , Paroxetina/toxicidade , Gravidez , Complicações na Gravidez/tratamento farmacológico , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Inibidores Seletivos de Recaptação de Serotonina/toxicidade
18.
Toxicol Appl Pharmacol ; 382: 114742, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31476325

RESUMO

Fluoxetine (FLX), the active ingredient in well-known therapeutic drugs such as Prozac, is highly prescribed worldwide to treat affective disorders even among pregnant women and adolescents. Given that FLX readily crosses the placenta, a fetus from a treated pregnant woman is potentially at risk from unintended effects of the chemical. Moreover, FLX reaches aquatic ecosystems at biologically active levels through sewage release, so fish may also be inadvertently affected. We previously demonstrated that FLX exposure to environmentally- (Low FLX Lineage; LFL) and human- (High FLX Lineage; HFL) relevant concentrations during the first 6 days of life in zebrafish (ZF; Danio rerio) reduced cortisol levels in the adults (F0), an effect that persisted across 3 consecutive unexposed generations (F1 to F3). Here, we show that the transcriptional profile of selected genes in the steroidogenesis pathway in the F0 whole-larvae varied in magnitude and direction in both FLX lineages, despite the same attenuated cortisol phenotype induced by both concentrations. We also observed an up-regulation in the transcript levels of some steroidogenic-related genes and a down-regulation of a gene involved in the inactivation of cortisol in the F3 HFL larvae. These findings on the transcript levels of the selected genes in the larvae from F0 and F3 suggest that specific coping mechanism(s) are activated in descendants to attempt to counteract the disruptive effects of FLX. Our data are cause for concern, given the increasing prescription rates of FLX and other antidepressants, and the potential long-term negative impacts on humans and aquatic organisms.


Assuntos
Fluoxetina/toxicidade , Regulação da Expressão Gênica no Desenvolvimento , Hidrocortisona/metabolismo , Larva/metabolismo , Estresse Psicológico/induzido quimicamente , Estresse Psicológico/metabolismo , Animais , Antidepressivos de Segunda Geração/toxicidade , Feminino , Hidrocortisona/genética , Larva/efeitos dos fármacos , Larva/genética , Masculino , Gravidez , Distribuição Aleatória , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Estresse Psicológico/genética , Poluentes Químicos da Água/toxicidade , Peixe-Zebra
19.
Pediatr Res ; 85(7): 1032-1040, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30739124

RESUMO

BACKGROUND: Approximately 1/3 of newborns exposed antenatally to selective serotonin reuptake inhibitors (SSRIs) exhibit poor neonatal adaptation. Although several potential mechanisms have been proposed, the actual mechanism has not been elucidated. METHODS: We investigated outcomes in neonatal lambs exposed prenatally or postnatally to fluoxetine (FX). Daily FX injections (50 mg) were given intravenously (i.v.) to five pregnant ewes via implanted catheters beginning at 131-132 days gestation (term = 147 days) for 2 weeks. In another group, lambs with implanted vascular catheters had sterile water (n = 9) or FX (1 mg/kg, n = 12) injected i.v. on ~postnatal day (PND) 4. RESULTS: Prenatal FX-exposed lambs (n = 7) were hyperactive during PND 4 to 14 and their heart rate variability (HRV) was significantly lower than in control lambs (n = 7) on PND 2. In contrast, arterial pressure, heart rate, electrocardiogram, arterial blood gases, pH, glucose, lactate, cortisol, and sleep-activity cycles were not altered following postnatal FX injection. CONCLUSION: This abnormal postnatal hyperactivity with antenatal FX exposure may reflect increased maturity in terms of locomotory activity. The results suggest that altered brain development may be involved in the poor neonatal adaptation in human infants exposed to FX in utero.


Assuntos
Fluoxetina/toxicidade , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Ovinos/embriologia , Animais , Animais Recém-Nascidos , Feminino , Fluoxetina/administração & dosagem , Frequência Cardíaca/efeitos dos fármacos , Exposição Materna , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem
20.
Arterioscler Thromb Vasc Biol ; 38(5): 1007-1019, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29567680

RESUMO

OBJECTIVE: Cardiovascular diseases and depression are the leading causes of disability in Western countries. Clinical data on potential cardiovascular effects of serotonin reuptake inhibitors (SSRIs), the most commonly used antidepressant drugs, are controversial. In addition to blocking serotonin reuptake transporter in the brain, SSRIs deplete the major peripheral serotonin (5-hydroxytryptamine [5-HT]) storage by inhibiting serotonin reuptake transporter-mediated uptake in platelets. In this study, we aimed to investigate the effect of chronic SSRI intake on the development of atherosclerosis. APPROACH AND RESULTS: Treatment of apolipoprotein E-deficient mice with the SSRI fluoxetine for 2, 4, or 16 weeks increased atherosclerotic lesion formation, with most pronounced effect during early plaque development. Intravital microscopy of carotid arteries revealed enhanced myeloid cell adhesion on fluoxetine treatment. Mechanistically, we found that fluoxetine augmented vascular permeability and increased chemokine-induced integrin-binding activity of circulating leukocytes. In vitro stimulation of murine blood demonstrated that fluoxetine, but not 5-HT, could directly promote ß1 and ß2 integrin activation provided C-C motif chemokine ligand 5 was also present. Similar effects were observed with the SSRI escitalopram. Enhanced C-C motif chemokine ligand 5-induced integrin activation by fluoxetine was also confirmed in a human neutrophil-like cell line. In contrast to the proatherogenic properties of fluoxetine, pharmacological inhibition of the peripheral 5-HT synthesizing enzyme tryptophan hydroxylase 1 did not promote atherosclerosis, suggesting that the proatherogenic effect of fluoxetine occurs independent of peripheral 5-HT depletion. CONCLUSIONS: SSRI intake may promote atherosclerosis and therefore potentially increase the risk for acute cardiovascular events by a mechanism that is independent of 5-HT depletion.


Assuntos
Aorta/efeitos dos fármacos , Doenças da Aorta/induzido quimicamente , Aterosclerose/induzido quimicamente , Artérias Carótidas/efeitos dos fármacos , Doenças das Artérias Carótidas/induzido quimicamente , Fluoxetina/toxicidade , Placa Aterosclerótica , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Animais , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/sangue , Doenças da Aorta/genética , Doenças da Aorta/patologia , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/patologia , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Antígenos CD18/sangue , Permeabilidade Capilar/efeitos dos fármacos , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/sangue , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/patologia , Adesão Celular/efeitos dos fármacos , Quimiocina CCL5/sangue , Modelos Animais de Doenças , Progressão da Doença , Esquema de Medicação , Fluoxetina/administração & dosagem , Células HEK293 , Células HL-60 , Humanos , Integrina beta1/sangue , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Serotonina/sangue , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Transdução de Sinais , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA