Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.262
Filtrar
1.
Sci Rep ; 14(1): 13762, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38877103

RESUMO

Selective Serotonin Reuptake Inhibitor (SSRI) therapy is common among perinatal populations for the treatment of mood disorders. Medications can affect diversity and composition of the gut microbiome, which plays a key role in modulating health. While previous studies have examined the effects of antidepressant exposure on the maternal gut microbiome, whether SSRI exposure affects the offspring gut microbiome is unknown. We investigated the effects of maternal fluoxetine exposure on the gut microbiome of maternal and offspring mice during pregnancy and lactation (embryonic day 10-lactation day 21; E10-L21). Stool samples collected on E17, L11, L15, and L21 were examined using 16S rRNA sequencing. Our results suggest that maternal fluoxetine exposure may result in decreased alpha diversity of the offspring gut microbiome in early life. Furthermore, we observed several genera-specific differences in the gut microbiome based on treatment, specifically of Turicibacter, Parasutterella, and Romboutsia. These findings support our understanding of gut health, as dysbiotic development of the gut microbiome has been associated with local and systemic health problems including gastrointestinal morbidities and interrupted growth patterns in infants. Future research should pursue study in human populations and those at high risk for gut microbial dysbiosis and intestinal injury.


Assuntos
Fluoxetina , Microbioma Gastrointestinal , Lactação , RNA Ribossômico 16S , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Feminino , Gravidez , Lactação/efeitos dos fármacos , Fluoxetina/farmacologia , Fluoxetina/efeitos adversos , Camundongos , RNA Ribossômico 16S/genética , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Inibidores Seletivos de Recaptação de Serotonina/efeitos adversos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Fezes/microbiologia , Exposição Materna/efeitos adversos , Bactérias/efeitos dos fármacos , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação
2.
Neurosci Lett ; 834: 137848, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38823510

RESUMO

Ketamine is a dissociative anesthetic that has been proposed to be a useful alternative in cases of a poor response to other treatments in patients with depression. Remarkably, beneficial clinical actions of ketamine are detected once its psychotropic actions disappear. Therefore, clinical actions may occur independently of dose. Most current studies focus on actions of ketamine on neurotrophic factors, but few studies have investigated actions of ketamine on neural structures for which actions of antidepressants have been previously explored. Lateral septal nucleus (LSN) stimulation reduces neural activity in the prelimbic cortex (PL) and infralimbic cortex (IL) subregions of the medial prefrontal cortex (mPFC). Fluoxetine increases inhibitory responsivity of the LSN-IL connection. In the present study, actions of an anesthetic dose of ketamine were compared with a high dose of fluoxetine on behavior and neural responsivity 24 h after drug administration. Fluoxetine reduced immobility in the forced swim test without changing locomotor activity in the open field test. Ketamine strongly decreased locomotor activity and did not produce changes in immobility. In another set of Wistar rats that received similar drug treatment regimens, the results indicated that LSN stimulation in saline-treated animals produced a long-lasting inhibitory afterdischarge in these mPFC subregions. Actions of ketamine on the LSN-mPFC connection reproduced actions of fluoxetine, consisting of accentuated inhibition of the LSN action on the mPFC. These findings suggest that independent of different actions on neurotransmission, the common final pathway of antidepressants lies in their actions on forebrain structures that are related to emotional regulation.


Assuntos
Fluoxetina , Ketamina , Córtex Pré-Frontal , Ratos Wistar , Núcleos Septais , Animais , Ketamina/farmacologia , Fluoxetina/farmacologia , Masculino , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Núcleos Septais/efeitos dos fármacos , Estimulação Elétrica
3.
Eur J Pharmacol ; 976: 176693, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38834095

RESUMO

ß-arrestin2 is a versatile protein for signaling transduction in brain physiology and pathology. Herein, we investigated the involvement of ß-arrestin2 in pharmacological effects of fluoxetine for depression. A chronic mild stress (CMS) model was established using wild-type (WT) and ß-arrestin2-/- mice. Behavioral results demonstrated that CMS mice showed increased immobility time in the tail suspension test and forced swimming test, elevated concentrations of pro-inflammatory factors in peripheral blood, increased expression of pyroptosis-related proteins, and increased co-labeling of glial fibrillary acidic protein and Caspase1 p10 in the hippocampus compared to the CON group. Treatment with fluoxetine (FLX) ameliorated these conditions. However, compared with the ß-arrestin2-/- CMS group, these results of the ß-arrestin2-/- CMS + FLX group showed no significant changes. These results suggested that the above effects of FLX could be eliminated by knocking out ß-arrestin2. Mass spectrometry implying that FLX promoted the binding of ß-arrestin2 to the NLRP2 inflammasome of depressed mice. Subsequently, the results of the cellular experiments suggested that the 5HT2B receptor antagonist may attenuate L-kynurenine + ATP-induced cell pyroptosis by attenuating NLRP2 binding to ß-arrestin2. We further found that the lack of ß-arrestin2 eliminated the anti-pyroptosis effect of fluoxetine. In conclusion, ß-arrestin2 is an essential protein for fluoxetine to alleviate pyroptosis in the hippocampal astrocytes of CMS mice. Mechanistically, we found that the 5-HT2BR-ß-arrestin2-NLRP2 axis is vital for maintaining the antidepressant effects of fluoxetine.


Assuntos
Antidepressivos , Astrócitos , Depressão , Modelos Animais de Doenças , Fluoxetina , Piroptose , Estresse Psicológico , beta-Arrestina 2 , Animais , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico , Piroptose/efeitos dos fármacos , beta-Arrestina 2/metabolismo , Camundongos , Depressão/tratamento farmacológico , Depressão/metabolismo , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/metabolismo , Masculino , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Camundongos Endogâmicos C57BL , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Camundongos Knockout , Comportamento Animal/efeitos dos fármacos , Inflamassomos/metabolismo , Doença Crônica
4.
Int Immunopharmacol ; 136: 112335, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-38815349

RESUMO

Chlorpyrifos (CPF) is a widely used organophosphate insecticide in agriculture and homes. Exposure to organophosphates is associated with neurotoxicity. Fluoxetine (FLX) is a selective serotonin reuptake inhibitor (SSRI) that is widely prescribed for depression and anxiety disorders. Studies have shown that FLX has neuroprotective, anti-inflammatory, antioxidant, and antiapoptotic effects. The molecular mechanisms underlying FLX are not fully understood. This work aimed to investigate the potential neuroprotective effect of FLX on CPF-induced neurotoxicity and the underlying molecular mechanisms involved. Thirty-two rats were randomly divided into four groups: (I) the vehicle control group; (II) the FLX-treated group (10 mg/kg/day for 28 days, p.o); (III) the CPF-treated group (10 mg/kg for 28 days); and (IV) the FLX+CPF group. FLX attenuated CPF-induced neuronal injury, as evidenced by a significant decrease in Aß and p-Tau levels and attenuation of cerebral and hippocampal histological abrasion injury induced by CPF. FLX ameliorated neuronal oxidative stress, effectively reduced MDA production, and restored SOD and GSH levels through the coactivation of the PPARγ and SIRT1 proteins. FLX counteracted the neuronal inflammation induced by CPF by decreasing MPO, NO, TNF-α, IL-1ß, and IL-6 levels by suppressing NF-κB and JAK1/STAT3 activation. The antioxidant and anti-inflammatory properties of FLX help to prevent CPF-induced neuronal intoxication.


Assuntos
Clorpirifos , Fluoxetina , Janus Quinase 1 , NF-kappa B , Fármacos Neuroprotetores , PPAR gama , Fator de Transcrição STAT3 , Transdução de Sinais , Sirtuína 1 , Animais , Fator de Transcrição STAT3/metabolismo , Sirtuína 1/metabolismo , NF-kappa B/metabolismo , PPAR gama/metabolismo , Janus Quinase 1/metabolismo , Masculino , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Clorpirifos/toxicidade , Ratos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Neurônios/efeitos dos fármacos , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Inseticidas/toxicidade , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Ratos Sprague-Dawley , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/patologia
5.
Nat Commun ; 15(1): 4434, 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38789416

RESUMO

Compulsive behaviors are a hallmark symptom of obsessive compulsive disorder (OCD). Striatal hyperactivity has been linked to compulsive behavior generation in correlative studies in humans and causal studies in rodents. However, the contribution of the two distinct striatal output populations to the generation and treatment of compulsive behavior is unknown. These populations of direct and indirect pathway-projecting spiny projection neurons (SPNs) have classically been thought to promote or suppress actions, respectively, leading to a long-held hypothesis that increased output of direct relative to indirect pathway promotes compulsive behavior. Contrary to this hypothesis, here we find that indirect pathway hyperactivity is associated with compulsive grooming in the Sapap3-knockout mouse model of OCD-relevant behavior. Furthermore, we show that suppression of indirect pathway activity using optogenetics or treatment with the first-line OCD pharmacotherapy fluoxetine is associated with reduced grooming in Sapap3-knockouts. Together, these findings highlight the striatal indirect pathway as a potential treatment target for compulsive behavior.


Assuntos
Comportamento Compulsivo , Modelos Animais de Doenças , Fluoxetina , Asseio Animal , Camundongos Knockout , Neurônios , Transtorno Obsessivo-Compulsivo , Optogenética , Animais , Transtorno Obsessivo-Compulsivo/fisiopatologia , Transtorno Obsessivo-Compulsivo/genética , Comportamento Compulsivo/fisiopatologia , Camundongos , Neurônios/metabolismo , Asseio Animal/fisiologia , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Masculino , Corpo Estriado/metabolismo , Comportamento Animal , Camundongos Endogâmicos C57BL , Feminino , Vias Neurais
6.
PLoS Negl Trop Dis ; 18(5): e0012199, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38776344

RESUMO

BACKGROUND: In Chagas disease (CD), a neglected tropical disease caused by the parasite Trypanosoma cruzi, the development of mental disorders such as anxiety, depression, and memory loss may be underpinned by social, psychological, and biological stressors. Here, we investigated biological factors underlying behavioral changes in a preclinical model of CD. METHODOLOGY/PRINCIPAL FINDINGS: In T. cruzi-infected C57BL/6 mice, a kinetic study (5 to 150 days postinfection, dpi) using standardized methods revealed a sequential onset of behavioral changes: reduced innate compulsive behavior, followed by anxiety and depressive-like behavior, ending with progressive memory impairments. Hence, T. cruzi-infected mice were treated (120 to 150 dpi) with 10 mg/Kg/day of the selective serotonin reuptake inhibitor fluoxetine (Fx), an antidepressant that favors neuroplasticity. Fx therapy reversed the innate compulsive behavior loss, anxiety, and depressive-like behavior while preventing or reversing memory deficits. Biochemical, histological, and parasitological analyses of the brain tissue showed increased levels of the neurotransmitters GABA/glutamate and lipid peroxidation products and decreased expression of brain-derived neurotrophic factor in the absence of neuroinflammation at 150 dpi. Fx therapy ameliorated the neurochemical changes and reduced parasite load in the brain tissue. Next, using the human U-87 MG astroglioma cell line, we found no direct effect of Fx on parasite load. Crucially, serotonin/5-HT (Ser/5-HT) promoted parasite uptake, an effect increased by prior stimulation with IFNγ and TNF but abrogated by Fx. Also, Fx blocked the cytokine-driven Ser/5-HT-promoted increase of nitric oxide and glutamate levels in infected cells. CONCLUSION/SIGNIFICANCE: We bring the first evidence of a sequential onset of behavioral changes in T. cruzi-infected mice. Fx therapy improves behavioral and biological changes and parasite control in the brain tissue. Moreover, in the central nervous system, cytokine-driven Ser/5-HT consumption may favor parasite persistence, disrupting neurotransmitter balance and promoting a neurotoxic environment likely contributing to behavioral and cognitive disorders.


Assuntos
Astrócitos , Doença de Chagas , Fluoxetina , Camundongos Endogâmicos C57BL , Serotonina , Trypanosoma cruzi , Animais , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico , Doença de Chagas/tratamento farmacológico , Doença de Chagas/psicologia , Trypanosoma cruzi/efeitos dos fármacos , Trypanosoma cruzi/fisiologia , Serotonina/metabolismo , Camundongos , Astrócitos/efeitos dos fármacos , Modelos Animais de Doenças , Encéfalo/efeitos dos fármacos , Encéfalo/parasitologia , Encéfalo/metabolismo , Comportamento Animal/efeitos dos fármacos , Masculino , Humanos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Cognição/efeitos dos fármacos , Depressão/tratamento farmacológico , Carga Parasitária , Ansiedade/tratamento farmacológico
7.
Phytomedicine ; 130: 155549, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-38810551

RESUMO

Premenstrual dysphoric disorder (PMDD) is a severe subtype of premenstrual syndrome in women of reproductive age, with its pathogenesis linked to the heightened sensitivity of type A γ -aminobutyric acid receptors (GABAAR) to neuroactive steroid hormone changes, particularly allopregnanolone (ALLO). While a low dose of fluoxetine, a classic selective serotonin reuptake inhibitor, is commonly used as a first-line drug to alleviate emotional disorders in PMDD in clinical settings, its mechanism of action is related to ALLO-GABAA receptor function. However, treating PMDD requires attention to both emotional and physical symptoms, such as pain sensitivity. This study aims to investigate the efficacy of ShuYu capsules, a traditional Chinese medicine, in simultaneously treating emotional and physical symptoms in a rat model of PMDD. Specifically, our focus centres on the midbrain periaqueductal grey (PAG), a region associated with emotion regulation and susceptibility to hyperalgesia. Considering the underlying mechanisms of ALLO-GABAA receptor function in the PAG region, we conducted a series of experiments to evaluate and define the effects of ShuYu capsules and uncover the relationship between the drug's efficacy and ALLO concentration fluctuations on GABAA receptor function in the PAG region. Our findings demonstrate that ShuYu capsules significantly improved oestrous cycle-dependant depression-like behaviour and reduced stress-induced hyperalgesia in rats with PMDD. Similar to the low dose of fluoxetine, ShuYu capsules targeted and mitigated the sharp decline in ALLO, rescued the upregulation of GABAAR subunit function, and activated PAG neurons in PMDD rats. The observed effects of ShuYu capsules suggest a central mechanism underlying PMDD symptoms, involving ALLO_GABAA receptor function in the PAG region. This study highlights the potential of traditional Chinese medicine in addressing both emotional and physical symptoms associated with PMDD, shedding light on novel therapeutic approaches for this condition.


Assuntos
Medicamentos de Ervas Chinesas , Pregnanolona , Transtorno Disfórico Pré-Menstrual , Ratos Sprague-Dawley , Receptores de GABA-A , Animais , Feminino , Medicamentos de Ervas Chinesas/farmacologia , Receptores de GABA-A/metabolismo , Pregnanolona/farmacologia , Transtorno Disfórico Pré-Menstrual/tratamento farmacológico , Ratos , Cápsulas , Modelos Animais de Doenças , Síndrome Pré-Menstrual/tratamento farmacológico , Fluoxetina/farmacologia
8.
Chem Biol Interact ; 397: 111041, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38719170

RESUMO

Abrocitinib is approved to treat moderate-to-severe atopic dermatitis and eliminated mainly through cytochrome P450 (CYP450) enzyme. Two commonly used antidepressants, amitriptyline and fluoxetine, could inhibit the activities of CYP2C19 and CYP3A4. In this study, we developed a new and quick ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method for quantitatively analyzing the plasma concentration of abrocitinib, and further investigated the effects of amitriptyline or fluoxetine on the pharmacokinetics of abrocitinib in rats. The selectivity, linearity, recovery, accuracy, precision, matrix effect and stability of UPLC-MS/MS assay were satisfied according to the United States Food and Drug Administration (FDA) and European Medicines Agency (EMA) guidelines. Our result showed that when co-administered with amitriptyline and fluoxetine, the CLz/F of abrocitinib was reduced by 44.4 % and 33.3 %, respectively, while the AUC(0-t) of abrocitinib was increased by 77.7 % and 49.4 %, respectively. It indicated that amitriptyline and fluoxetine could significantly increase the plasma concentration of abrocitinib in rats. Thus, dose adjustment of abrocitinib may be required when it is combined with amitriptyline or fluoxetine in ongoing clinical practice.


Assuntos
Amitriptilina , Fluoxetina , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem , Animais , Fluoxetina/farmacocinética , Fluoxetina/farmacologia , Ratos , Masculino , Amitriptilina/farmacocinética , Antidepressivos/farmacocinética , Antidepressivos/farmacologia , Cromatografia Líquida de Alta Pressão , Interações Medicamentosas , Pirimidinas/farmacocinética , Pirimidinas/farmacologia , Pirimidinas/sangue
9.
Artigo em Inglês | MEDLINE | ID: mdl-38692472

RESUMO

Stress exposure can lead to post-traumatic stress disorder (PTSD) in male and female rats. Social-Single Prolonged Stress (SPS) protocol has been considered a potential PTSD model. This study aimed to pharmacologically validate the Social-SPS as a PTSD model in male and female rats. Male and female Wistar rats (60-day-old) were exposed to Social-SPS protocol and treated with fluoxetine (10 mg/Kg) or saline solution intraperitoneally 24 h before euthanasia. Two cohorts of animals were used; for cohort 1, male and female rats were still undisturbed until day 7 post-Social-SPS exposure, underwent locomotor and conditioned fear behaviors, and were euthanized on day 9. Animals of cohort 2 were subjected to the same protocol but were re-exposed to contextual fear behavior on day 14. Results showed that fluoxetine-treated rats gained less body weight than control and Social-SPS in both sexes. Social-SPS effectively increased the freezing time in male and female rats on day eight but not on day fourteen. Fluoxetine blocked the increase of freezing in male and female rats on day 8. Different mechanisms for fear behavior were observed in males, such as Social-SPS increased levels of glucocorticoid receptors and Beclin-1 in the amygdala. Social-SPS was shown to increase the levels of NMDA2A, GluR-1, PSD-95, and CAMKII in the amygdala of female rats. No alterations were observed in the amygdala of rats on day fourteen. The study revealed that Social-SPS is a potential PTSD protocol applicable to both male and female rats.


Assuntos
Tonsila do Cerebelo , Medo , Fluoxetina , Ratos Wistar , Estresse Psicológico , Animais , Masculino , Feminino , Medo/efeitos dos fármacos , Medo/fisiologia , Fluoxetina/farmacologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Estresse Psicológico/metabolismo , Ratos , Modelos Animais de Doenças , Transtornos de Estresse Pós-Traumáticos/metabolismo , Transtornos de Estresse Pós-Traumáticos/psicologia , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Proteína 4 Homóloga a Disks-Large , Receptores de AMPA
10.
Dev Psychobiol ; 66(5): e22501, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38807259

RESUMO

Selective serotonin reuptake inhibitors, such as fluoxetine (Prozac), are commonly prescribed pharmacotherapies for anxiety. Fluoxetine may be a useful adjunct because it can reduce the expression of learned fear in adult rodents. This effect is associated with altered expression of perineuronal nets (PNNs) in the amygdala and hippocampus, two brain regions that regulate fear. However, it is unknown whether fluoxetine has similar effects in adolescents. Here, we investigated the effect of fluoxetine exposure during adolescence or adulthood on context fear memory and PNNs in the basolateral amygdala (BLA), the CA1 subregion of the hippocampus, and the medial prefrontal cortex in rats. Fluoxetine impaired context fear memory in adults but not in adolescents. Further, fluoxetine increased the number of parvalbumin (PV)-expressing neurons surrounded by a PNN in the BLA and CA1, but not in the medial prefrontal cortex, at both ages. Contrary to previous reports, fluoxetine did not shift the percentage of PNNs toward non-PV cells in either the BLA or CA1 in the adults, or adolescents. These findings demonstrate that fluoxetine differentially affects fear memory in adolescent and adult rats but does not appear to have age-specific effects on PNNs.


Assuntos
Medo , Fluoxetina , Memória , Córtex Pré-Frontal , Inibidores Seletivos de Recaptação de Serotonina , Fluoxetina/farmacologia , Fluoxetina/administração & dosagem , Animais , Medo/efeitos dos fármacos , Medo/fisiologia , Masculino , Ratos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Córtex Pré-Frontal/efeitos dos fármacos , Memória/efeitos dos fármacos , Memória/fisiologia , Fatores Etários , Ratos Sprague-Dawley , Parvalbuminas/metabolismo , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Complexo Nuclear Basolateral da Amígdala/metabolismo , Região CA1 Hipocampal/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos
11.
Biol Pharm Bull ; 47(5): 946-954, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38735732

RESUMO

There is accumulating evidence that selective serotonin reuptake inhibitors (SSRIs), clinically used as antidepressants, have a beneficial effect on inflammatory diseases such as coronavirus disease 2019 (COVID-19). We previously compared the inhibitory effects of five U.S. Food and Drug Administration (FDA)-approved SSRIs on the production of an inflammatory cytokine, interleukin-6 (IL-6), and concluded that fluoxetine (FLX) showed the most potent anti-inflammatory activity. Here, we investigated the structure-activity relationship of FLX for anti-inflammatory activity towards J774.1 murine macrophages. FLX suppressed IL-6 production induced by the TLR3 agonist polyinosinic-polycytidylic acid (poly(I : C)) with an IC50 of 4.76 µM. A derivative of FLX containing chlorine instead of the methylamino group lacked activity, suggesting that the methylamino group is important for the anti-inflammatory activity. FLX derivatives bearing an N-propyl or N-(pyridin-3-yl)methyl group in place of the N-methyl group exhibited almost the same activity as FLX. Other derivatives showed weaker activity, and the N-phenyl and N-(4-trifluoromethyl)benzyl derivatives were inactive. The chlorine-containing derivative also lacked inhibitory activity against TLR9- or TLR4-mediated IL-6 production. These derivatives showed similar structure-activity relationships for TLR3- and TLR9-mediated inflammatory responses. However, the activities of all amino group-containing derivatives against the TLR4-mediated inflammatory response were equal to or higher than the activity of FLX. These results indicate that the substituent at the nitrogen atom in FLX strongly influences the anti-inflammatory effect.


Assuntos
Anti-Inflamatórios , Fluoxetina , Interleucina-6 , Relação Estrutura-Atividade , Animais , Fluoxetina/farmacologia , Camundongos , Interleucina-6/metabolismo , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Linhagem Celular , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Citocinas/metabolismo , Receptor 3 Toll-Like/metabolismo , Poli I-C/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/química , Inflamação/tratamento farmacológico
12.
Genes (Basel) ; 15(4)2024 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-38674386

RESUMO

Down syndrome (DS) is the most common form of inherited intellectual disability caused by trisomy of chromosome 21, presenting with intellectual impairment, craniofacial abnormalities, cardiac defects, and gastrointestinal disorders. The Ts65Dn mouse model replicates many abnormalities of DS. We hypothesized that investigation of the cerebral cortex of fluoxetine-treated trisomic mice may provide proteomic signatures that identify therapeutic targets for DS. Subcellular fractionation of synaptosomes from cerebral cortices of age- and brain-area-matched samples from fluoxetine-treated vs. water-treated trisomic and euploid male mice were subjected to HPLC-tandem mass spectrometry. Analysis of the data revealed enrichment of trisomic risk genes that participate in regulation of synaptic vesicular traffic, pre-synaptic and post-synaptic development, and mitochondrial energy pathways during early brain development. Proteomic analysis of trisomic synaptic fractions revealed significant downregulation of proteins involved in synaptic vesicular traffic, including vesicular endocytosis (CLTA, CLTB, CLTC), synaptic assembly and maturation (EXOC1, EXOC3, EXOC8), anterograde axonal transport (EXOC1), neurotransmitter transport to PSD (SACM1L), endosomal-lysosomal acidification (ROGDI, DMXL2), and synaptic signaling (NRXN1, HIP1, ITSN1, YWHAG). Additionally, trisomic proteomes revealed upregulation of several trafficking proteins, involved in vesicular exocytosis (Rab5B), synapse elimination (UBE3A), scission of endocytosis (DBN1), transport of ER in dendritic spines (MYO5A), presynaptic activity-dependent bulk endocytosis (FMR1), and NMDA receptor activity (GRIN2A). Chronic fluoxetine treatment of Ts65Dn mice rescued synaptic vesicular abnormalities and prevented abnormal proteomic changes in adult Ts65Dn mice, pointing to therapeutic targets for potential treatment of DS.


Assuntos
Síndrome de Down , Fluoxetina , Proteômica , Vesículas Sinápticas , Animais , Fluoxetina/farmacologia , Camundongos , Síndrome de Down/metabolismo , Síndrome de Down/tratamento farmacológico , Síndrome de Down/genética , Síndrome de Down/patologia , Masculino , Proteômica/métodos , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/efeitos dos fármacos , Modelos Animais de Doenças , Proteoma/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/efeitos dos fármacos , Sinaptossomos/metabolismo , Sinaptossomos/efeitos dos fármacos , Trissomia/genética
13.
Behav Pharmacol ; 35(4): 227-238, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38651981

RESUMO

We have previously reported that two inhibitors of an E3 ligase S-phase kinase-associated protein 2 (Skp2), SMIP004 and C1, have an antidepressant-like effect in non-stressed and chronically stressed mice. This prompted us to ask whether other Skp2 inhibitors could also have an antidepressant effect. Here, we used NSC689857, another Skp2 inhibitor, to investigate this hypothesis. The results showed that administration of NSC689857 (5 mg/kg) produced an antidepressant-like effect in a time-dependent manner in non-stressed male mice, which started 8 days after drug administration. Dose-dependent analysis showed that administration of 5 and 10 mg/kg, but not 1 mg/kg, of NSC689857 produced antidepressant-like effects in both non-stressed male and female mice. Administration of NSC689857 (5 mg/kg) also induced antidepressant-like effects in non-stressed male mice when administered three times within 24 h (24, 5, and 1 h before testing) but not when administered acutely (1 h before testing). In addition, NSC689857 and fluoxetine coadministration produced additive antidepressant-like effects in non-stressed male mice. These effects of NSC689857 were not associated with the changes in locomotor activity. Administration of NSC689857 (5 mg/kg) also attenuated depression-like behaviors in male mice induced by chronic social defeat stress, suggesting therapeutic potential of NSC689857 in depression. Overall, these results suggest that NSC689857 is capable of exerting antidepressant-like effects in both non-stressed and chronically stressed mice.


Assuntos
Antidepressivos , Benzotiepinas , Relação Dose-Resposta a Droga , Proteínas Quinases Associadas a Fase S , Animais , Feminino , Masculino , Camundongos , Antidepressivos/farmacologia , Comportamento Animal/efeitos dos fármacos , Depressão/tratamento farmacológico , Modelos Animais de Doenças , Fluoxetina/farmacologia , Proteínas Quinases Associadas a Fase S/antagonistas & inibidores , Estresse Psicológico/tratamento farmacológico
14.
Eur J Clin Pharmacol ; 80(7): 983-1016, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38558317

RESUMO

Brain-derived neurotrophic factor (BDNF) dysfunction is one of the most important mechanisms underlying depression. It seems that selective serotonin reuptake inhibitors (SSRIs) improve depression via affecting BDNF level. In this systematic review, for the first time, we aimed to review the effect of three SSRIs including fluoxetine, escitalopram, and sertraline, on both depression and BDNF level in preclinical and clinical studies. PubMed electronic database was searched, and 193 articles were included in this study. After reviewing all manuscripts, only one important difference was found: subjects. We found that SSRIs induce different effects in animals vs. humans. Preclinical studies showed many controversial effects, while human studies showed only two effects: improvement of depression, with or without the improvement of BDNF. However, most studies used chronic SSRIs treatment, while acute SSRIs were not effectively used and evaluated. In conclusion, it seems that SSRIs are reliable antidepressants, and the improvement effect of SSRIs on depression is not dependent to BDNF level (at least in human studies).


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Depressão , Fluoxetina , Inibidores Seletivos de Recaptação de Serotonina , Sertralina , Humanos , Fator Neurotrófico Derivado do Encéfalo/sangue , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Sertralina/farmacologia , Sertralina/uso terapêutico , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Fluoxetina/farmacologia , Animais , Depressão/tratamento farmacológico , Escitalopram/farmacologia , Escitalopram/uso terapêutico , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico
15.
Epilepsia ; 65(6): 1791-1800, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38593237

RESUMO

OBJECTIVE: Sudden unexpected death in epilepsy (SUDEP) is an underestimated complication of epilepsy. Previous studies have demonstrated that enhancement of serotonergic neurotransmission suppresses seizure-induced sudden death in evoked seizure models. However, it is unclear whether elevated serotonin (5-HT) function will prevent spontaneous seizure-induced mortality (SSIM), which is characteristic of human SUDEP. We examined the effects of 5-HT-enhancing agents that act by three different pharmacological mechanisms on SSIM in Dravet mice, which exhibit a high incidence of SUDEP, modeling human Dravet syndrome. METHODS: Dravet mice of both sexes were evaluated for spontaneous seizure characterization and changes in SSIM incidence induced by agents that enhance 5-HT-mediated neurotransmission. Fluoxetine (a selective 5-HT reuptake inhibitor), fenfluramine (a 5-HT releaser and agonist), SR 57227 (a specific 5-HT3 receptor agonist), or saline (vehicle) was intraperitoneally administered over an 8-day period in Dravet mice, and the effect of these treatments on SSIM was examined. RESULTS: Spontaneous seizures in Dravet mice generally progressed from wild running to tonic seizures with or without SSIM. Fluoxetine at 30 mg/kg, but not at 20 or 5 mg/kg, significantly reduced SSIM compared with the vehicle control. Fenfluramine at 1-10 mg/kg, but not .2 mg/kg, fully protected Dravet mice from SSIM, with all mice surviving. Compared with the vehicle control, SR 57227 at 20 mg/kg, but not at 10 or 5 mg/kg, significantly lowered SSIM. The effect of these drugs on SSIM was independent of sex. SIGNIFICANCE: Our data demonstrate that elevating serotonergic function by fluoxetine, fenfluramine, or SR 57227 significantly reduces or eliminates SSIM in Dravet mice in a sex-independent manner. These findings suggest that deficits in serotonergic neurotransmission likely play an important role in the pathogenesis of SSIM, and fluoxetine and fenfluramine, which are US Food and Drug Administration-approved medications, may potentially prevent SUDEP in at-risk patients.


Assuntos
Epilepsias Mioclônicas , Fenfluramina , Fluoxetina , Convulsões , Inibidores Seletivos de Recaptação de Serotonina , Serotonina , Animais , Camundongos , Masculino , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico , Feminino , Epilepsias Mioclônicas/tratamento farmacológico , Fenfluramina/farmacologia , Convulsões/tratamento farmacológico , Convulsões/prevenção & controle , Convulsões/etiologia , Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Modelos Animais de Doenças , Morte Súbita Inesperada na Epilepsia/prevenção & controle , Agonistas do Receptor de Serotonina/farmacologia , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.1/genética
16.
Exp Gerontol ; 190: 112432, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38614224

RESUMO

The beneficial effect of social interaction in mitigating the incidence of post-stroke depression (PSD) and ameliorating depressive symptoms has been consistently demonstrated through preclinical and clinical studies. However, the underlying relationship with oxytocin requires further investigation. In light of this, the present study aimed to explore the protective effect of pair housing on the development of PSD and the potential relationship with oxytocin receptors. The PSD model was induced by middle cerebral artery occlusion (MCAO) for 50 min, followed by 4-week isolated housing and restrained stress. Subsequently, each mouse in the pair-housing group (PH) was pair-housed with an isosexual healthy partner. Another group was continuously administrated fluoxetine (10 mg/Kg, i.p, once a day) for 3 weeks. To elucidate the potential role of oxytocin, we subjected pair-housed PSD mice to treatment with an oxytocin receptor (OXTR) antagonist (L368,889) (5 mg/Kg, i.p, once a day) for 3 weeks. At 31 to 32 days after MCAO, anxiety- and depressive-like behaviors were assessed using sucrose consumption, forced swim test, and tail-suspension test. The results showed that pair housing significantly improved post-stroke depression to an extent comparable to that of fluoxetine treatment. Furthermore, pair housing significantly decreased corticosterone in serum, increasing OXT mRNA expression in the hypothalamus. Treatment with L368,889 essentially reversed the effect of pair housing, with no discernible sex differences apart from changes in body weight. Pair housing increased hippocampal serotonin (5-HT), but treatment with L368,889 had no significant impact. Additionally, pair housing effectively reduced the number of reactive astrocytes and increased Nissl's body in the cortex and hippocampal CA3 regions. Correspondingly, treatment with L368,889 significantly reversed the changes in the Nissl's body and reactive astrocytes. Moreover, pair housing downregulated mRNA levels of TNF-α, IL-1ß, and IL-6 in the cortex caused by PSD, which was also reversed by treatment with L368,889. In conclusion, pair housing protects against the development of PSD depending on OXT and OXTR in the brain, with no significant divergence based on sex. These findings provide valuable insights into the potential of social interaction and oxytocin as therapeutic targets for PSD. Further research into the underlying mechanisms of these effects may contribute to the development of novel treatments for PSD.


Assuntos
Canfanos , Depressão , Modelos Animais de Doenças , Fluoxetina , Piperazinas , Receptores de Ocitocina , Animais , Receptores de Ocitocina/metabolismo , Masculino , Depressão/etiologia , Depressão/metabolismo , Camundongos , Fluoxetina/farmacologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/psicologia , Abrigo para Animais , Ocitocina/farmacologia , Ocitocina/metabolismo , Camundongos Endogâmicos C57BL , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/psicologia , Comportamento Animal/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos
17.
Pharmacol Biochem Behav ; 240: 173779, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38688436

RESUMO

The use of a selective serotonin reuptake inhibitor fluoxetine in depression during pregnancy and the postpartum period might increase the risk of affective disorders and cognitive symptoms in progeny. In animal models, maternal exposure to fluoxetine throughout gestation and lactation negatively affects the behavior of the offspring. Little is known about the effects of maternal fluoxetine on synaptic transmission and plasticity in the offspring cerebral cortex. During pregnancy and lactation C57BL/6J mouse dams received fluoxetine (7.5 mg/kg/day) with drinking water. Female offspring mice received intraperitoneal injections of the selective 5-HT7 receptor antagonist SB 269970 (2.5 mg/kg) for 7 days. Whole-cell and field potential electrophysiological recordings were performed in the medial prefrontal cortex (mPFC) ex vivo brain slices. Perinatal exposure to fluoxetine resulted in decreased field potentials and impaired long-term potentiation (LTP) in layer II/III of the mPFC of female young adult offspring. Neither the intrinsic excitability nor spontaneous excitatory postsynaptic currents were altered in layer II/III mPFC pyramidal neurons. In mPFC slices obtained from fluoxetine-treated mice that were administered SB 269970 both field potentials and LTP magnitude were restored and did not differ from controls. Treatment of fluoxetine-exposed mice with a selective 5-HT7 receptor antagonist, SB 269970, normalizes synaptic transmission and restores the potential for plasticity in the mPFC of mice exposed in utero and postnatally to fluoxetine.


Assuntos
Fluoxetina , Camundongos Endogâmicos C57BL , Plasticidade Neuronal , Fenóis , Córtex Pré-Frontal , Efeitos Tardios da Exposição Pré-Natal , Receptores de Serotonina , Sulfonamidas , Animais , Fluoxetina/farmacologia , Feminino , Camundongos , Córtex Pré-Frontal/efeitos dos fármacos , Receptores de Serotonina/efeitos dos fármacos , Receptores de Serotonina/metabolismo , Gravidez , Plasticidade Neuronal/efeitos dos fármacos , Fenóis/farmacologia , Sulfonamidas/farmacologia , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Antagonistas da Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Potenciação de Longa Duração/efeitos dos fármacos
18.
Chemosphere ; 355: 141851, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38579950

RESUMO

Fish have common neurotransmitter pathways with humans, exhibiting a significant degree of conservation and homology. Thus, exposure to fluoxetine makes fish potentially susceptible to biochemical and physiological changes, similarly to what is observed in humans. Over the years, several studies demonstrated the potential effects of fluoxetine on different fish species and at different levels of biological organization. However, the effects of parental exposure to unexposed offspring remain largely unknown. The consequences of 15-day parental exposure to relevant concentrations of fluoxetine (100 and 1000 ng/L) were assessed on offspring using zebrafish as a model organism. Parental exposure resulted in offspring early hatching, non-inflation of the swimming bladder, increased malformation frequency, decreased heart rate and blood flow, and reduced growth. Additionally, a significant behavioral impairment was also found (reduced startle response, basal locomotor activity, and altered non-associative learning during early stages and a negative geotaxis and scototaxis, reduced thigmotaxis, and anti-social behavior at later life stages). These behavior alterations are consistent with decreased anxiety, a significant increase in the expression of the monoaminergic genes slc6a4a (sert), slc6a3 (dat), slc18a2 (vmat2), mao, tph1a, and th2, and altered levels of monoaminergic neurotransmitters. Alterations in behavior, expression of monoaminergic genes, and neurotransmitter levels persisted until offspring adulthood. Given the high conservation of neuronal pathways between fish and humans, data show the possibility of potential transgenerational and multigenerational effects of pharmaceuticals' exposure. These results reinforce the need for transgenerational and multigenerational studies in fish, under realistic scenarios, to provide realistic insights into the impact of these pharmaceuticals.


Assuntos
Perciformes , Poluentes Químicos da Água , Animais , Humanos , Adulto , Peixe-Zebra/metabolismo , Fluoxetina/farmacologia , Larva , Antidepressivos/farmacologia , Perciformes/metabolismo , Neurotransmissores/metabolismo , Preparações Farmacêuticas/metabolismo , Poluentes Químicos da Água/metabolismo
19.
Viruses ; 16(4)2024 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-38675888

RESUMO

The pandemic caused by SARS-CoV-2 is still a major health problem. Newly emerging variants and long-COVID-19 represent a challenge for the global health system. In particular, individuals in developing countries with insufficient health care need easily accessible, affordable and effective treatments of COVID-19. Previous studies have demonstrated the efficacy of functional inhibitors of acid sphingomyelinase against infections with various viruses, including early variants of SARS-CoV-2. This work investigated whether the acid sphingomyelinase inhibitors fluoxetine and sertraline, usually used as antidepressant molecules in clinical practice, can inhibit the replication of the former and recently emerged SARS-CoV-2 variants in vitro. Fluoxetine and sertraline potently inhibited the infection with pseudotyped virus-like particles and SARS-CoV-2 variants D614G, alpha, delta, omicron BA.1 and omicron BA.5. These results highlight fluoxetine and sertraline as priority candidates for large-scale phase 3 clinical trials at different stages of SARS-CoV-2 infections, either alone or in combination with other medications.


Assuntos
Antivirais , COVID-19 , Fluoxetina , SARS-CoV-2 , Sertralina , Replicação Viral , SARS-CoV-2/efeitos dos fármacos , Sertralina/farmacologia , Fluoxetina/farmacologia , Replicação Viral/efeitos dos fármacos , Humanos , Antivirais/farmacologia , Chlorocebus aethiops , Células Vero , COVID-19/virologia , Animais , Tratamento Farmacológico da COVID-19
20.
Artigo em Inglês | MEDLINE | ID: mdl-38552774

RESUMO

Alzheimer's disease (AD) and depression are inflammatory pathologies, leading to increased inflammatory response and neurotoxicity. Therefore, this study aimed to evaluate the effect of the treatment with fluoxetine and/or galantamine and/or donepezil on the levels of proinflammatory and anti-inflammatory cytokines in a mixed animal model of depression and dementia. Adult male Wistar rats underwent chronic mild stress (CMS) protocol for 40 days and were subjected to stereotaxic surgery for intra-hippocampal administration of amyloid-beta (Aꞵ) peptide or artificial cerebrospinal fluid (ACSF) to mimic the dementia animal model. On the 42nd day, animals were treated with water, galantamine, donepezil, and/or fluoxetine, orally for 17 days. On the 57th and 58th days, the Splash and Y-maze tests for behavior analysis were performed. The frontal cortex and hippocampus were used to analyze the tumor necrosis factor alfa (TNF-α), interleukin 1 beta (IL-1ꞵ), IL-6, and IL-10 levels. The results of this study show that animals subjected to CMS and administration of Aꞵ had anhedonia, cognitive impairment, increased TNF-α and IL-1ꞵ levels in the frontal cortex, and reduced IL-10 levels in the hippocampus. All treatment groups were able to reverse the cognitive impairment. Only donepezil did not decrease the TNF-α levels in the hippocampus. Fluoxetine + galantamine and fluoxetine + donepezil reversed the anhedonia. Fluoxetine reversed the anhedonia and IL-1ꞵ levels in the frontal cortex. In addition, fluoxetine + donepezil reversed the reduction of IL-10 levels in the hippocampus. The results indicate a pathophysiological interaction between AD and depression, and the association of medications in the future may be a possible therapeutic strategy to reduce inflammation, especially the fluoxetine-associated treatments.


Assuntos
Demência , Depressão , Modelos Animais de Doenças , Donepezila , Fluoxetina , Galantamina , Hipocampo , Ratos Wistar , Animais , Masculino , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico , Donepezila/farmacologia , Donepezila/uso terapêutico , Ratos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Demência/tratamento farmacológico , Depressão/tratamento farmacológico , Galantamina/farmacologia , Galantamina/uso terapêutico , Citocinas/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Estresse Psicológico/complicações , Peptídeos beta-Amiloides/metabolismo , Anedonia/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...