Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
BMC Pharmacol Toxicol ; 25(1): 51, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-39152503

RESUMO

BACKGROUND: Fluoxetine is present in breast milk, yet it is unclear to what extent it, or its active metabolite, norfluoxetine, reaches the brain of the infant and what the effects of such exposure on neurobiological processes are. We therefore aimed to quantify the concentration of passively administered fluoxetine and norfluoxetine in the whole brains of exposed Flinders sensitive line (FSL) offspring and establish their influence on serotonergic function and redox status. METHODS: Adult FSL dams received fluoxetine (10 mg/kg/day), or placebo for fourteen days, beginning on postpartum day 04. Offspring were passively exposed to fluoxetine until postnatal day 18 and euthanized on postnatal day 22. Whole brain fluoxetine, norfluoxetine, serotonin (5-HT), 5-hydroxyindoleacetic acid (5-HIAA), and reduced (GSH) and oxidized glutathione (GSSG) concentrations were measured via liquid chromatography-mass spectrometry (LC-MS) analysis. RESULTS: Whole-brain serotonin and 5-hydroxyindoleacetic acid concentrations, and serotonin turnover (5-HIAA/5-HT) were comparable between strains. Treatment-naïve FSL rats had lower GSH and higher GSSG whole-brain concentrations, relative to FRL controls, and an overall decreased GSH/GSSG ratio. Passively administered fluoxetine resulted in undetectable whole-brain concentrations, while norfluoxetine averaged 41.28 ± 6.47 ng/g. Serotonin turnover of FSL rats was unaffected by passively administered fluoxetine, while redox status (GSH/GSSG) was decreased. CONCLUSION: Our findings confirm that passively administered fluoxetine reaches the infant brain in the form of norfluoxetine and may manipulate processes of oxidative stress regulation. Further studies into the long-term bio-behavioural effects are however needed to effectively inform breast feeding mothers on the safety of antidepressant-use.


Assuntos
Encéfalo , Fluoxetina , Inibidores Seletivos de Recaptação de Serotonina , Serotonina , Animais , Fluoxetina/análogos & derivados , Fluoxetina/farmacologia , Serotonina/metabolismo , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Feminino , Ratos , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Masculino , Gravidez , Glutationa/metabolismo
2.
Anal Bioanal Chem ; 412(21): 5113-5123, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32162085

RESUMO

A rapid, simple, and sensitive technique for the quantitative detection of fluoxetine and norfluoxetine enantiomers in biological fluids was developed based on the combination of field-amplified sample stacking (FASS)-related capillary electrophoresis (CE) with ultrasound-assisted dispersive liquid-liquid microextraction (UA-DLLME). The extraction efficiency of UA-DLLME was strongly related to extraction time, salt concentration, type of extraction and dispersion solvents, and volume of extraction and dispersion solvents. The extracted fluoxetine and norfluoxetine enantiomers in a mixture of 50% methanol and 50% deionized water were efficiently stacked using FASS and then separated using cyclodextrin-modified CE. Under optimal conditions of FASS (chiral selector, 3 mM trimethyl-ß-cyclodextrin; and background electrolyte, 100 mM phosphate buffer) and UA-DLLME (extraction solvent, 200 µL of acetone; and dispersed solvent, 50 µL of C2H2Cl4 in 1 mL of the sample solution), the obtained enrichment factors of fluoxetine and norfluoxetine enantiomers reached approximately 2000. The linear ranges for the quantification of fluoxetine and norfluoxetine enantiomers were 0.3-150 and 0.6-150 nM, respectively. The relative standard deviations in peak areas and migration time for four analytes were less than 3.3% and 6.3%, respectively. The proposed system provided limits of detection (signal-to-noise ratio of 3) for four analytes corresponding to 0.1 nM. The precision and accuracy for urine and serum samples were less than 6.8 and 8.3%, respectively. These findings suggested that the proposed system exhibited a high potential for the reliable determination of fluoxetine and norfluoxetine enantiomers in clinical samples. Graphical abstract.


Assuntos
Líquidos Corporais/química , Eletroforese Capilar/métodos , Fluoxetina/análogos & derivados , Fluoxetina/análise , Microextração em Fase Líquida/métodos , Inibidores Seletivos de Recaptação de Serotonina/análise , Sonicação , Fluoxetina/sangue , Fluoxetina/urina , Humanos , Limite de Detecção , Inibidores Seletivos de Recaptação de Serotonina/sangue , Inibidores Seletivos de Recaptação de Serotonina/urina , Espectrofotometria Ultravioleta/métodos , Estereoisomerismo
3.
Behav Brain Res ; 378: 112266, 2020 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-31580915

RESUMO

Psychostimulants are highly effective cognitive-enhancing therapeutics yet have a significant potential for abuse and addiction. While psychostimulants likely exert their rewarding and addictive properties through dopamine transporter (DAT) inhibition, the mechanisms of their procognitive effects are less certain. By one prevalent view, psychostimulants exert their procognitive effects exclusively through norepinephrine transporter (NET) inhibition, however increasing evidence suggests that DAT also plays a critical role in their cognitive-enhancing properties, including long-term memory enhancement. The present experiments test the hypothesis that combined strong NET and weak DAT inhibition will mimic the fear memory-enhancing but not the addiction-related effects of psychostimulants in mice. We examined the effects of the high affinity NET inhibitors atomoxetine or nisoxetine and the low affinity DAT inhibitor bupropion, either alone or in combination, on short- and long-term memory of Pavlovian fear conditioning. We also examined the addiction-related effects of combined strong NET and weak DAT inhibition using conditioned place preference and a locomotor activity test. While atomoxetine or nisoxetine alone enhanced short-term fear memory, the addition of bupropion was required to significantly enhance long-term fear memory. Additionally, combined atomoxetine and bupropion did not produce substantial motor stimulation or place preference. These findings suggest that combining strong NET and weak DAT inhibition could lead to the development of a highly effective cognitive enhancer that lacks the potential for addiction.


Assuntos
Cloridrato de Atomoxetina/farmacologia , Comportamento Animal/efeitos dos fármacos , Bupropiona/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Dopamina/antagonistas & inibidores , Medo/efeitos dos fármacos , Fluoxetina/análogos & derivados , Memória de Longo Prazo/efeitos dos fármacos , Inibidores da Captação de Neurotransmissores/farmacologia , Nootrópicos/farmacologia , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/antagonistas & inibidores , Animais , Cloridrato de Atomoxetina/administração & dosagem , Bupropiona/administração & dosagem , Estimulantes do Sistema Nervoso Central/administração & dosagem , Quimioterapia Combinada , Feminino , Fluoxetina/administração & dosagem , Fluoxetina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibidores da Captação de Neurotransmissores/administração & dosagem , Nootrópicos/administração & dosagem
4.
J Steroid Biochem Mol Biol ; 195: 105470, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31509772

RESUMO

Serotonin reuptake inhibitors (SRIs) are currently the main molecules prescribed to pregnant women that suffer from depression. Placental cells are exposed to SRIs via maternal blood, and we have previously shown that SRIs alter feto-placental steroidogenesis in an in vitro co-culture model. More specifically, serotonin (5-HT) regulates the estrogen biosynthetic enzyme aromatase (cytochrome P450 19; CYP19), which is disrupted by fluoxetine and its active metabolite norfluoxetine in BeWo choriocarcinoma cells. Based on molecular simulations, the present study illustrates that the SRIs fluoxetine, norfluoxetine, paroxetine, sertraline, citalopram and venlafaxine exhibit binding affinity for the active-site pocket of CYP19, suggesting potential competitive inhibition. Using BeWo cells and primary villous trophoblast cells isolated from normal term placentas, we compared the effects of the SRIs on CYP19 activity. We observed that paroxetine and sertraline induce aromatase activity in BeWo cells, while venlafaxine, fluoxetine, paroxetine and sertraline decrease aromatase activity in primary villous trophoblast. The effects of paroxetine and sertraline in primary villous trophoblasts were observed at the lower doses tested. We also showed that 5-HT and the 5-HT2A receptor agonist 2,5-dimethoxy-4-iodoamphetamine (DOI) induced CYP19 activity. An increase in phosphorylation of serine and tyrosine and a decrease in threonine phosphorylation of CYP19 was also associated with DOI treatment. Our results contribute to better understanding how 5-HT and SRIs interact with CYP19 and may affect estrogen production. Moreover, this study suggests that alteration of placental 5-HT levels due to depression and/or SRI treatment during pregnancy may be associated with disruption of placental estrogen production.


Assuntos
Aromatase/metabolismo , Placenta/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Serotonina/farmacologia , Células Cultivadas , Citalopram/farmacologia , Feminino , Fluoxetina/análogos & derivados , Fluoxetina/farmacologia , Humanos , Simulação de Acoplamento Molecular , Paroxetina/farmacologia , Placenta/metabolismo , Gravidez , Sertralina/farmacologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Cloridrato de Venlafaxina/farmacologia
5.
Behav Pharmacol ; 30(6): 463-470, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30724802

RESUMO

Pain is a significant public health problem, and assessment of pain-related impairment of behavior is a key clinical indicator and treatment target. Similar to opioids and NSAIDs, dopamine (DA) transporter inhibitors block pain-related depression of intracranial self-stimulation (ICSS) in rats. The primary goal of the present study was to determine if the effects of monoamine uptake inhibitors on pain-related depression of ICSS in rats extend to an assay of pain-related depression of nesting in mice. We hypothesized that the DA transporter-selective uptake inhibitor bupropion would block depression of nesting behavior produced by intraperitoneal injection of lactic acid, whereas selective serotonin transporter-selective citalopram, norepinephrine transporter-selective nisoxetine, and the mixed action selective serotonin transporter/norepinephrine transporter inhibitor milnacipran would be ineffective. Effects of the NSAID ketoprofen were also obtained to facilitate interpretation of the effects of the monoamine uptake inhibitors. Consistent with previous findings, ketoprofen blocked pain-related depression of nesting. In contrast, none of the monoamine uptake inhibitors blocked pain-related depression of nesting, although they all blocked pain-related stimulation of stretching. Unlike findings from studies of pain-related depression of ICSS, these results do not support consideration of DA uptake inhibitors for treatment of pain-related depression of behavior.


Assuntos
Comportamento Animal/efeitos dos fármacos , Comportamento de Nidação/efeitos dos fármacos , Dor/tratamento farmacológico , Analgésicos Opioides/farmacologia , Animais , Bupropiona/farmacologia , Citalopram/farmacologia , Condicionamento Operante/efeitos dos fármacos , Inibidores da Captação de Dopamina/farmacologia , Fluoxetina/análogos & derivados , Fluoxetina/farmacologia , Cetoprofeno/farmacologia , Ácido Láctico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Milnaciprano/farmacologia , Receptores Opioides kappa/metabolismo , Receptores Opioides kappa/fisiologia , Autoestimulação/efeitos dos fármacos
6.
Placenta ; 72-73: 62-73, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30501883

RESUMO

INTRODUCTION: Between 2 and 10% of pregnant women are treated with selective serotonin-reuptake inhibitors (SSRIs) for depression. The extravillous trophoblasts (evTBs), which migrate and invade maternal tissues, are crucial for embryo implantation and remodeling of maternal spiral arteries. Poor migration/invasion of evTBs can cause serious pregnancy complications, yet the effects of SSRIs on these processes has never been studied. To determine the effects of five SSRIs (fluoxetine, norfluoxetine, citalopram, sertraline and venlafaxine) on migration/invasion, we used JEG-3 and HIPEC cells as evTB models. METHODS: Cells were treated with increasing concentrations (0.03-10 µM) of SSRIs. Cell proliferation was monitored using an impedance-based system and cell cycle by flow cytometry. Migration was determined using a scratch test, and metalloproteinase (MMP) activities, by zymography. Invasion markers were determined by RT-qPCR. RESULTS: Fluoxetine and sertraline (10 µM) significantly decreased cell proliferation by 94% and by 100%, respectively, in JEG-3 cells, and by 58.6% and 100%, respectively, in HIPEC cells. Norfluoxetine increased MMP-9 activity in JEG-3 cells by 2.0% at 0.03 µM and by 43.9% at 3 µM, but decreased MMP-9 activity in HIPEC cells by 63.7% at 3 µM. Sertraline at 0.03 µM increased mRNA level of TIMP-1 in JEG-3 cells by 36% and that of ADAM-10 by 85% and 115% at 0.3 and 3 µM, respectively. In HIPEC cells, venlafaxine at 0.03 and 0.3 µM, increased ADAM-10 mRNA levels by 156% and 167%, respectively. DISCUSSION: This study shows that SSRIs may affect evTBs homeostasis at therapeutic levels and provides guidance for future research.


Assuntos
Inibidores Seletivos de Recaptação de Serotonina/efeitos adversos , Trofoblastos/efeitos dos fármacos , Proteína ADAM10/genética , Secretases da Proteína Precursora do Amiloide/genética , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Coriocarcinoma , Feminino , Fluoxetina/efeitos adversos , Fluoxetina/análogos & derivados , Expressão Gênica/efeitos dos fármacos , Humanos , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Metaloproteinase 9 da Matriz/metabolismo , Proteínas de Membrana/genética , Modelos Biológicos , Gravidez , RNA Mensageiro/análise , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Sertralina/efeitos adversos , Inibidor Tecidual de Metaloproteinase-1/genética , Trofoblastos/fisiologia , Cloridrato de Venlafaxina/efeitos adversos
7.
Chemosphere ; 186: 958-967, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28830067

RESUMO

The disposition and metabolism of fluoxetine in the European shore crab and the Dungeness crab were assessed. Crabs received intracardiac doses of either 0.13 µg/kg or 0.5 mg/kg fluoxetine, respectively. In addition, fluoxetine was administered to Metacarcinus cancer by oral gavage at 7.8 mg/kg. The distribution of fluoxetine was quantified in haemolymph and digestive gland for both crabs, as well as brain, muscle, and testis of Carcinus maenas, over 12 days. The metabolite norfluoxetine, was also measured in C. maenas. Fluoxetine was mainly found in lipid rich tissues. Distribution coefficients increased for digestive gland until three days after fluoxetine administration and then decreased until the end of the observations. The highest distribution coefficients were obtained for brain. Norfluoxetine displayed continuously high levels in digestive gland and brain. The strong decrease in fluoxetine and the concomitant increase in norfluoxetine demonstrates that decapod crustaceans metabolise fluoxetine into the more biologically active norfluoxetine. Fluoxetine levels in the haemolymph of M. cancer declined within 20 h, but showed a second peak 25 h later, suggesting remobilisation from tissues sequestering the compound. The steady state volume distribution and the total body clearance of fluoxetine were high, consistent with high diffusion of fluoxetine into the peripheral tissues and biotransformation as an important elimination pathway. Oral administration of fluoxetine prolonged its half-life in M. cancer, but bioavailability was low. These results confirm the high distribution into nervous tissue, extensive biotransformation into the highly active norfluoxetine and a half-life similar to that observed in vertebrates.


Assuntos
Braquiúros/metabolismo , Fluoxetina/metabolismo , Fluoxetina/farmacocinética , Animais , Biotransformação , Fluoxetina/análogos & derivados , Fluoxetina/análise , Fluoxetina/toxicidade , Meia-Vida , Distribuição Tecidual , Toxicocinética
8.
Mol Cell Endocrinol ; 442: 32-39, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27890559

RESUMO

The effects of fluoxetine, one of the most prescribed selective serotonin-reuptake inhibitors (SSRIs) during pregnancy, and its active metabolite norfluoxetine were studied on placental aromatase (CYP19) and feto-placental steroidogenesis. Fluoxetine did not alter estrogen secretion in co-culture of fetal-like adrenocortical (H295R) and trophoblast-like (BeWo) cells used as a model of the feto-placental unit, although it induced CYP19 activity, apparently mediated by the serotonin (5-HT)2A receptor/PKC signaling pathway. Norfluoxetine decreased estrogen secretion in the feto-placental co-culture and competitively inhibited catalytic CYP19 activity in BeWo cells. Decreased serotonin transporter (SERT) activity in the co-culture was comparable to 17ß-estradiol treatment of BeWo cells. This work shows that the complex interaction of fluoxetine and norfluoxetine with placental estrogen production, involves 5-HT-dependent and -independent mechanisms. Considering the crucial role of estrogens during pregnancy, our results raise concern about the impact of SSRI treatment on placental function and fetal health.


Assuntos
Estrogênios/metabolismo , Fluoxetina/análogos & derivados , Fluoxetina/farmacologia , Placenta/efeitos dos fármacos , Aromatase/metabolismo , Linhagem Celular Tumoral , Técnicas de Cocultura/métodos , Estradiol/metabolismo , Feminino , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Placenta/metabolismo , Gravidez , Proteína Quinase C/metabolismo , Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Transdução de Sinais/efeitos dos fármacos
9.
Hum Psychopharmacol ; 31(3): 178-84, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27018372

RESUMO

OBJECTIVE: Only 30% of major depressive disorder (MDD) patients achieve complete remission with a serotonergic antidepressant (selective serotonin reuptake inhibitor). We investigated the potential of serotonin (5-HT) and interleukin-6 (IL-6) to serve as functional biomarkers of fluoxetine response. METHODS: Serum IL-6 and 5-HT were measured in 73 MDD patients (39 responders and 34 non-responders) pre- and 6 weeks post-treatment and in 44 normal controls with ELISA. Fluoxetine and norfluoxetine were measured using LC MS/MS. RESULTS: IL-6 levels were significantly higher in MDD patients when compared with controls (p < 0.01), and 5-HT levels were significantly lower in non-responders compared with controls (p = 0.0131). Pre- and post-treatment levels of both biomarkers individually and in combination did not significantly differ between responders and non-responders. Area under the receiver operating characteristics curve for the biomarkers was 0.5. Significant correlation was seen between the percentage change in IL-6 and percentage change in Hamilton Rating Scale for Depression score in responders. Fluoxetine and norfluoxetine concentrations were not significantly different in responders and non-responders, and there was no correlation between fluoxetine concentrations and percentage reduction in 5-HT from week 0 to 6. CONCLUSION: 5-HT and IL-6 may not serve as useful markers of response to fluoxetine because of inconsistent results across different studies. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Transtorno Depressivo Maior/tratamento farmacológico , Fluoxetina/uso terapêutico , Interleucina-6/sangue , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Serotonina/sangue , Adolescente , Adulto , Idoso , Biomarcadores/sangue , Estudos de Casos e Controles , Cromatografia Líquida , Transtorno Depressivo Maior/fisiopatologia , Ensaio de Imunoadsorção Enzimática , Feminino , Fluoxetina/análogos & derivados , Fluoxetina/farmacocinética , Humanos , Masculino , Pessoa de Meia-Idade , Escalas de Graduação Psiquiátrica , Inibidores Seletivos de Recaptação de Serotonina/farmacocinética , Espectrometria de Massas em Tandem , Resultado do Tratamento , Adulto Jovem
10.
J UOEH ; 37(1): 33-42, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25787100

RESUMO

The norepinephrine transporter is selectively expressed in noradrenergic nerve terminals, where it can exert spatial and temporal control over the action of norepinephrine. The norepinephrine transporter mediates the termination of neurotransmission via the reuptake of norepinephrine released into the extracellular milieu. In the present brief review, we report our recent studies about the effects of various pharmacological agents such as fasudil, nicotine, pentazocine, ketamine and genistein on norepinephrine transporter function.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Genisteína/farmacologia , Ketamina/farmacologia , Nicotina/farmacologia , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/fisiologia , Pentazocina/farmacologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Neurônios Adrenérgicos/metabolismo , Animais , Células Cultivadas , Fluoxetina/análogos & derivados , Fluoxetina/metabolismo , Humanos , Terminações Nervosas/metabolismo , Norepinefrina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/fisiologia , Transmissão Sináptica/genética
11.
Neural Plast ; 2014: 123026, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24757568

RESUMO

Selective serotonin reuptake inhibitor medications are one of the most common treatments for mood disorders. In humans, these medications are taken orally, usually once per day. Unfortunately, administration of antidepressant medications in rodent models is often through injection, oral gavage, or minipump implant, all relatively stressful procedures. The aim of the present study was to investigate how administration of the commonly used SSRI, fluoxetine, via a wafer cookie, compares to fluoxetine administration using an osmotic minipump, with regards to serum drug levels and hippocampal plasticity. For this experiment, adult female Sprague-Dawley rats were divided over the two administration methods: (1) cookie and (2) osmotic minipump and three fluoxetine treatment doses: 0, 5, or 10 mg/kg/day. Results show that a fluoxetine dose of 5 mg/kg/day, but not 10 mg/kg/day, results in comparable serum levels of fluoxetine and its active metabolite norfluoxetine between the two administration methods. Furthermore, minipump administration of fluoxetine resulted in higher levels of cell proliferation in the granule cell layer (GCL) at a 5 mg dose compared to a 10 mg dose. Synaptophysin expression in the GCL, but not CA3, was significantly lower after fluoxetine treatment, regardless of administration method. These data suggest that the administration method and dose of fluoxetine can differentially affect hippocampal plasticity in the adult female rat.


Assuntos
Fluoxetina/farmacologia , Hipocampo/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Animais , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Região CA3 Hipocampal/citologia , Região CA3 Hipocampal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Estradiol/sangue , Feminino , Fluoxetina/administração & dosagem , Fluoxetina/análogos & derivados , Fluoxetina/sangue , Bombas de Infusão Implantáveis , Antígeno Ki-67/análise , Ratos , Ratos Sprague-Dawley , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Sinaptofisina/análise , Sinaptofisina/metabolismo
12.
Chirality ; 26(3): 166-73, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24464553

RESUMO

Fluoxetine (FLX) is one of the most widely prescribed selective serotonin reuptake inhibitors. Although FLX is used as racemate in the clinic, the clinical pharmacokinetics of FLX and its N-demethylation metabolite norfluoxetine (NFLX) show obvious cytochrome P450 (CYP) polymorphism dependency and exhibit marked stereoselectivity. However, the kinetic profiles of CYP variants to FLX remain unclear. In the present study, some variants of human CYP2C8, CYP2C9, and CYP2D6 were first expressed in insect cells, and their catalytic roles with respect to FLX enantiomers were then investigated. CYP2C8.4 and CYP2C9.10 showed significantly lower activity and CYP2C8.3 showed significantly higher activity toward both R- and S-FLX compared with the wildtype, while CYP2C9.3, CYP2C9.13, and CYP2C9.16 showed significantly lower activity only toward R-FLX. Five CYP2C9 variants and CYP2D6.1 exhibited significantly stereoselective kinetic profiles prior to R-FLX, and CYP2C8.3 showed a slight stereoselectivity. Interestingly, obvious substrate inhibition was observed in the CYP2C9 wildtype and its three variants only in the case of R-FLX. Together, these findings suggest that CYP2C9 and CYP2D6 polymorphism may play an important role in the clearance of FLX and also in the stereoselective kinetic profiles of FLX enantiomers.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Fluoxetina/análogos & derivados , Polimorfismo Genético , Alelos , Citocromo P-450 CYP2C8 , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/metabolismo , Fluoxetina/química , Fluoxetina/metabolismo , Humanos , Cinética , Metilação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estereoisomerismo
13.
Pharmacol Rep ; 65(2): 350-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23744419

RESUMO

BACKGROUND: Although nisoxetine has been shown to elicit infiltrative cutaneous local anesthesia, the inhibition of voltage-gated Na(+) channels by nisoxetine has not been reported. The aim of this study was to evaluate the effect of nisoxetine on Na(+) currents and its efficacy on spinal anesthesia. METHODS: In in vitro studies, the voltage-clamp method was employed to examine whether nisoxetine blocked Na(+) currents in mouse neuroblastoma N2A cells. RESULTS: Mepivacaine showed concentration- and state-dependent effect on tonic blockade of voltage-gated Na(+) currents (IC50 of 3.7 and 74.2 µM at holding potentials of -70 and -100 mV, respectively). Nisoxetine was more potent (IC50 of 1.6 and 28.6 µM at holding potentials of -70 and -100 mV, respectively). In in vivo studies, after rats were intrathecally injected with nisoxetine and mepivacaine, the dose-response curves were constructed. Nisoxetine acted like local anesthetic mepivacaine and induced spinal anesthesia with a more sensory-selective action (p < 0.05) over motor blockade in a dose-related fashion. Intrathecal 5% dextrose (vehicle) produced no spinal anesthesia. On the 50% effective dose (ED50) basis, nisoxetine elicited more potent spinal anesthesia than did mepivacaine (p < 0.05). CONCLUSIONS: Our results showed that nisoxetine displayed a more potent and prolonged spinal anesthesia with a more sensory/nociceptive-selective action over motor blockade, compared with mepivacaine. The local anesthetic effect of nisoxetine could be probably due to the suppression of Na(+) currents.


Assuntos
Raquianestesia/métodos , Anestésicos Locais/farmacologia , Fluoxetina/análogos & derivados , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Anestésicos Locais/administração & dosagem , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Fluoxetina/administração & dosagem , Fluoxetina/farmacologia , Concentração Inibidora 50 , Injeções Espinhais , Masculino , Mepivacaína/administração & dosagem , Mepivacaína/farmacologia , Camundongos , Neuroblastoma/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Bloqueadores do Canal de Sódio Disparado por Voltagem/administração & dosagem , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Canais de Sódio Disparados por Voltagem/metabolismo
14.
Neurosci Lett ; 541: 184-9, 2013 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-23485735

RESUMO

α2-Adrenergic agonists simulate norepinephrine (NE) action on α2 receptors of sympathetic neurons to mediate feedback inhibition of NE release. These agents are used as valuable adjuncts for management of hypertension and for anesthesia. Their action, equivalent to NE on α2 adrenergic receptors, raises the question whether α2 agonists may also target NE transporters (NETs), another major control mechanism for noradrenergic neurotransmission. We thus investigated the effect of α2 agonists on transport of the NE analog, (131)I-metaiodobenzylguanidine (MIBG). Results from this investigation showed that xylazine and dexmedetomidine dose-dependently blocked [(3)H]nisoxetine binding in neuron-like SK-N-SH cells. Furthermore, the agents acutely suppressed cellular MIBG uptake in a dose-dependent manner. This effect was uninfluenced by the α2 antagonist yohimbine, but was completely reversed by drug removal. There was no change in membrane NET density by the agents. Moreover, saturation analysis showed that xylazine and dexmedetomidine significantly increased Km without affecting Vmax, indicating competitive inhibition of MIBG transport. Thus, the α2 adrenergic agonists xylazine and dexmedetomidine, acutely suppress NET function through competitive inhibition of substrate transport, likely by direct interaction on a region that over-laps with the nisoxetine binding site.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Dexmedetomidina/farmacologia , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/antagonistas & inibidores , Norepinefrina/metabolismo , Xilazina/farmacologia , 3-Iodobenzilguanidina/metabolismo , Linhagem Celular Tumoral , Fluoxetina/análogos & derivados , Fluoxetina/metabolismo , Humanos , Radioisótopos do Iodo , Ligação Proteica , Receptores Adrenérgicos alfa 2/metabolismo
15.
Neurochem Res ; 38(7): 1303-14, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23525969

RESUMO

The thiol reagent N-ethylmaleimide (NEM) is known to inhibit irreversibly ligand binding by the norepinephrine transporter (NET), while the simultaneous presence of NET substrates or ligands protects from this inhibition. Therefore, cysteine residues located within the substrate binding pocket of the NET were assumed to play an important role in ligand binding. To examine which (if any) of the 10 cysteines (Cys) of the human (h) NET might be involved in transport and/or binding function, we mutated all hNET cysteines to alanine. Using transfected HEK293 cells we studied NEM effects on the hNET with respect to [(3)H]nisoxetine binding. Two cysteines (Cys176 and Cys185) within the extracellular loop of the NET have been proposed to form a disulfide bond. We could demonstrate that this is of crucial importance as corresponding hNET mutants, in which these cysteines have been replaced, showed a lack of plasma membrane expression. However, due to their oxidized state in the native NET protein, Cys176 and Cys185 may not be targets for NEM. All other Cys-to-Ala hNET mutants were fully active and showed no change in inhibition of [(3)H]nisoxetine binding by NEM. These observations clearly exclude cysteines as being involved in hNET ligand binding. Since NEM also interacts with histidin (His), we mutated all 13 histidins of the hNET to alanine and examined the NET mutants in functional and binding assays. His222 within the large extracellular loop of the transporter was identified as an interaction partner of NEM since in the corresponding hNET mutant NEM exhibited a significantly reduced inhibitory potency. Furthermore, we could show that histidins in position 296, 370 and 372 are important for nisoxetine binding, while His220, 441, 598 and 599 are crucial for plasma membrane expression of the hNET.


Assuntos
Cisteína/metabolismo , Histidina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Etilmaleimida/metabolismo , Fluoxetina/análogos & derivados , Fluoxetina/metabolismo , Células HEK293 , Humanos , Mutagênese Sítio-Dirigida , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/química , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/genética , Ensaio Radioligante , Frações Subcelulares/metabolismo
16.
J Obes ; 2013: 457047, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23431425

RESUMO

Central noradrenergic pathways are involved in feeding and cardiovascular control, physiological processes altered by obesity. The present studies determined how high-fat feeding and body weight gain alter the sensitivity to the feeding suppression and neural activation to a selective norepinephrine reuptake inhibitor, nisoxetine. Acute administration of nisoxetine (saline: 0, 3, 10, and 30 mg/kg; i.p.) resulted in a dose-dependent reduction in the 24 h refeeding response in male Sprague Dawley rats maintained on standard chow. In a similar fashion, nisoxetine resulted in reductions in blood pressure and a compensatory increase in heart rate. From these studies, the 3 mg/kg dose was subthreshold. In a separate experiment, however, 10 wk exposure to a high-fat diet (60% fat) resulted in weight gain and significant feeding suppression following administration of nisoxetine (3 mg/kg) compared with animals fed a control diet (10% fat). Nisoxetine (3 mg/kg) also resulted in greater neural activation, as measured by c-Fos immunohistochemistry, in the arcuate nucleus of the hypothalamus in animals exposed to the high-fat diet. Such data indicate acute nisoxetine doses that suppress food intake can impact cardiovascular measures. It also suggests that the feeding suppression to a low-dose nisoxetine is enhanced as a result of high-fat diet and weight gain.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Ingestão de Alimentos/efeitos dos fármacos , Fluoxetina/análogos & derivados , Norepinefrina/antagonistas & inibidores , Norepinefrina/fisiologia , Animais , Regulação do Apetite/efeitos dos fármacos , Regulação do Apetite/fisiologia , Núcleo Arqueado do Hipotálamo/química , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fluoxetina/administração & dosagem , Frequência Cardíaca/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Proteínas Proto-Oncogênicas c-fos/análise , Ratos , Ratos Sprague-Dawley , Aumento de Peso/efeitos dos fármacos
17.
J Pharmacol Sci ; 121(2): 138-47, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23370666

RESUMO

(±)-Pentazocine (PTZ), a non-narcotic analgesic, is used for the clinical management of moderate to severe pain. To study the effect of PTZ on the descending noradrenergic inhibitory system, in the present study we examined the effect of [(3)H]norepinephrine (NE) uptake by cultured bovine adrenal medullary cells and human neuroblastoma SK-N-SH cells. (-)-PTZ and (+)-PTZ inhibited [(3)H]NE uptake by adrenal medullary cells in a concentration-dependent (3-100 µM) manner. Eadie-Hofstee analysis of [(3)H]NE uptake showed that both PTZs caused a significant decrease in the V(max) with little change in the apparent K(m), suggesting non-competitive inhibition. Nor-Binaltorphimine and BD-1047, κ-opioid and σ-receptor antagonists, respectively, did not affect the inhibition of [(3)H]NE uptake induced by (-)-PTZ and (+)-PTZ, respectively. PTZs suppressed specific [(3)H]nisoxetine binding to intact SK-N-SH cells, but not directly to the plasma membranes isolated from the bovine adrenal medulla. Scatchard analysis of [(3)H]nisoxetine binding to SK-N-SH cells revealed that PTZs reduced the B(max) without changing the apparent K(d). Western blot analysis showed a decrease in biotinylated cell-surface NE transporter (NET) expression after the treatment with (-)-PTZ. These findings suggest that PTZ inhibits the NET function by reducing the amount of NET in the cell surface membranes through an opioid and σ-receptor-independent pathway.


Assuntos
Analgésicos Opioides/farmacologia , Antagonistas de Entorpecentes/farmacologia , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/antagonistas & inibidores , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Pentazocina/farmacologia , Medula Suprarrenal/diagnóstico por imagem , Medula Suprarrenal/efeitos dos fármacos , Medula Suprarrenal/metabolismo , Animais , Bovinos , Linhagem Celular , Membrana Celular/diagnóstico por imagem , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Etilenodiaminas/farmacologia , Fluoxetina/análogos & derivados , Fluoxetina/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Norepinefrina/metabolismo , Cintilografia
18.
Neurochem Res ; 38(4): 694-704, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23338678

RESUMO

Pro-inflammatory cytokines have been implicated in the precipitation of depression and related disorders, and the antidepressant sensitive serotonin transporter (SERT) may be a major target for immune regulation in these disorders. Here, we focus on astrocytes, a major class of immune competent cells in the brain, to examine the effects of pro-longed treatment with tumor necrosis factor-alpha (TNF-α) on SERT activity. We first established that high-affinity serotonin uptake into C6 glioma cells occurs through a SERT-dependent mechanism. Functional SERT expression is also confirmed for primary astrocytes. In both cell types, exposure to TNF-α resulted in a dose- and time-dependent increase in SERT-mediated 5-HT uptake, which was sustained for at least 48 h post-stimulation. Further analysis in primary astrocytes revealed that TNF-α enhanced the transport capacity (Vmax) of SERT-specific 5-HT uptake, suggesting enhanced transporter expression, consistent with our observation of an increase in SERT mRNA levels. We confirmed that in both, primary astrocytes and C6 glioma cells, treatment with TNF-α activates the p38 mitogen-activated protein kinase (MAPK) signaling pathway. Pre-treatment with the p38 MAPK inhibitor SB203580 attenuated the TNF-α mediated stimulation of 5-HT transport in both, C6 glioma and primary astrocytes. In summary, we show that SERT gene expression and activity in astrocytes is subject to regulation by TNF-α, an effect that is at least in part dependent on p38 MAPK activation.


Assuntos
Astrócitos/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/biossíntese , Fator de Necrose Tumoral alfa/farmacologia , Astrócitos/efeitos dos fármacos , Linhagem Celular Tumoral , Citalopram/farmacologia , Fluoxetina/análogos & derivados , Fluoxetina/farmacologia , Glioma/metabolismo , Humanos , Imidazóis/farmacologia , Paroxetina/farmacologia , Piridinas/farmacologia , Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Neuropsychopharmacology ; 37(11): 2522-30, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22781838

RESUMO

Prepulse inhibition (PPI) deficits are among the most reproducible phenotypic markers found in schizophrenic patients. We recently reported that nisoxetine, a selective norepinephrine transporter (NET) inhibitor, reversed the PPI deficits that have been identified in dopamine transporter (DAT) knockout (KO) mice. However, the mechanisms underlying nisoxetine-induced PPI recovery in DAT KO mice were unclear in previous experiments. To clarify these mechanisms, PPI was tested after microinjections of nisoxetine into the medial prefrontal cortex (mPFc) or nucleus accumbens (NAc) in wildtype (WT) and DAT KO mice. c-Fos immunohistochemistry provided an indicator of neural activation. Multiple-fluorescent-labeling procedures and the retrograde tracer fluorogold were employed to identify nisoxetine-activated neurons and circuits. Systemic nisoxetine activated the mPFc, the NAc shell, the basolateral amygdala, and the subiculum. Infusions of nisoxetine into the mPFc reversed PPI deficits in DAT KO mice, but produced no changes in WT mice, while infusion of nisoxetine into the NAc had no effect on PPI in both WT and DAT KO mice. Experiments using multiple-fluorescent labeling/fluorogold revealed that nisoxetine activates presumed glutamatergic pyramidal cells that project from the mPFc to the NAc. Activated glutamatergic projections from the mPFc to the NAc appear to have substantial roles in the ability of a NET inhibitor to normalize PPI deficits in DAT KO. Thus, this data suggest that selective NET inhibitors such as nisoxetine might improve information processing deficits in schizophrenia via regulation of cortico-subcortical neuromodulation.


Assuntos
Encéfalo/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/deficiência , Regulação da Expressão Gênica/genética , Inibição Psicológica , Reflexo de Sobressalto/genética , Estimulação Acústica/efeitos adversos , Análise de Variância , Animais , Encéfalo/efeitos dos fármacos , Vias de Administração de Medicamentos , Fluoxetina/análogos & derivados , Fluoxetina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato Descarboxilase/metabolismo , Glutaminase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Neurotransmissores/metabolismo , Inibidores da Captação de Neurotransmissores/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Reflexo de Sobressalto/efeitos dos fármacos , Fatores Sexuais , Estilbamidinas
20.
Pharmacopsychiatry ; 45(2): 72-6, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22086744

RESUMO

INTRODUCTION: Information about therapeutic serum levels of fluoxetine (FLX) and its major metabolite norfluoxetine (NORFLX) in children and adolescents is scarce. METHODS: Therapeutic drug monitoring (TDM) of FLX was routinely performed in 71 subjects treated for a major depressive disorder (MDD) (10-60 mg/d FLX, median: 20 mg/d). Correlations between serum concentration and dosage, age, gender, smoking habits and adverse events were analysed. RESULTS: Serum concentrations of the active moiety (FLX + NORFLX) ranged from 21 to 613 ng/mL (mean concentration of 213 ± 118 ng/mL, median: 185 ng/mL). High inter-individual variability in serum concentrations of the active moiety of FLX at each dosage level was observed and no relationship between serum concentration and clinical outcome was found. Apart from smoking, none of the factors tested had a significant eff ect on the serum concentration. DISCUSSION: It was shown that serum concentrations of the active moiety of FLX in children and adolescents seem to be similar to those in adults, with a high level of inter-individual variation. The proportion of patients who showed benefits from treatment with a dose of 20 mg/d FLX was high.


Assuntos
Transtorno Depressivo Maior/tratamento farmacológico , Monitoramento de Medicamentos/estatística & dados numéricos , Fluoxetina/farmacocinética , Fluoxetina/uso terapêutico , Inibidores Seletivos de Recaptação de Serotonina/farmacocinética , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Adolescente , Fatores Etários , Criança , Estudos de Coortes , Transtorno Depressivo Maior/sangue , Transtorno Depressivo Maior/psicologia , Relação Dose-Resposta a Droga , Monitoramento de Medicamentos/métodos , Estudos de Viabilidade , Feminino , Fluoxetina/efeitos adversos , Fluoxetina/análogos & derivados , Fluoxetina/sangue , Humanos , Masculino , Inibidores Seletivos de Recaptação de Serotonina/efeitos adversos , Inibidores Seletivos de Recaptação de Serotonina/sangue , Caracteres Sexuais , Fumar/psicologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA