Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.278
Filtrar
1.
BMC Pharmacol Toxicol ; 25(1): 51, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39152503

RESUMEN

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.


Asunto(s)
Encéfalo , Fluoxetina , Inhibidores Selectivos de la Recaptación de Serotonina , Serotonina , Animales , Fluoxetina/análogos & derivados , Fluoxetina/farmacología , Serotonina/metabolismo , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Femenino , Ratas , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Antioxidantes/metabolismo , Antioxidantes/farmacología , Masculino , Embarazo , Glutatión/metabolismo
2.
Cells ; 13(15)2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39120293

RESUMEN

Major depressive disorder (MDD) is a complex and devastating illness that affects people of all ages. Despite the large use of antidepressants in current medical practice, neither their mechanisms of action nor the aetiology of MDD are completely understood. Experimental evidence supports the involvement of Parvalbumin-positive GABAergic neurons (PV-neurons) in the pathogenesis of MDD. DLX5 and DLX6 (DLX5/6) encode two homeodomain transcription factors involved in cortical GABAergic differentiation and function. In the mouse, the level of expression of these genes is correlated with the cortical density of PV-neurons and with anxiety-like behaviours. The same genomic region generates the lncRNA DLX6-AS1, which, in humans, participates in the GABAergic regulatory module downregulated in schizophrenia and ASD. Here, we show that the expression levels of Dlx5/6 in the adult mouse brain are correlated with the immobility time in the forced swim test, which is used to measure depressive-like behaviours. We show that the administration of the antidepressant fluoxetine (Flx) to normal mice induces, within 24 h, a rapid and stable reduction in Dlx5, Dlx6 and Dlx6-AS1 expression in the cerebral cortex through the activation of the TrkB-CREB pathway. Experimental Dlx5 overexpression counteracts the antidepressant effects induced by Flx treatment. Our findings show that one of the short-term effects of Flx administration is the reduction in Dlx5/6 expression in GABAergic neurons, which, in turn, has direct consequences on PV expression and on behavioural profiles. Variants in the DLX5/6 regulatory network could be implicated in the predisposition to depression and in the variability of patients' response to antidepressant treatment.


Asunto(s)
Antidepresivos , Corteza Cerebral , Fluoxetina , Neuronas GABAérgicas , Proteínas de Homeodominio , Receptor trkB , Animales , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Fluoxetina/farmacología , Fluoxetina/uso terapéutico , Ratones , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Corteza Cerebral/metabolismo , Receptor trkB/metabolismo , Receptor trkB/genética , Masculino , Transducción de Señal/efectos de los fármacos , Ratones Endogámicos C57BL , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/metabolismo , Trastorno Depresivo Mayor/patología , Trastorno Depresivo Mayor/genética
3.
Behav Pharmacol ; 35(6): 314-326, 2024 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-39094014

RESUMEN

Depression is a common mood disorder and many patients do not respond to conventional pharmacotherapy or experience a variety of adverse effects. This work proposed that riparin I (RIP I) and riparin II (RIP II) present neuroprotective effects through modulation of astrocytes and microglia, resulting in the reversal of depressive-like behaviors. To verify our hypothesis and clarify the pathways underlying the effect of RIP I and RIP II on neuroinflammation, we used the chronic unpredictable mild stress (CUMS) depression model in mice. Male Swiss mice were exposed to stressors for 28 days. From 15 th to the 22 nd day, the animals received RIP I or RIP II (50 mg/kg) or fluoxetine (FLU, 10 mg/kg) or vehicle, by gavage. On the 29 th day, behavioral tests were performed. Expressions of microglia (ionized calcium-binding adaptor molecule-1 - Iba-1) and astrocyte (glial fibrillary acidic protein - GFAP) markers and levels of cytokines tumor necrosis factor alfa (TNF-α) and interleukin 1 beta (IL-1ß) were measured in the hippocampus. CUMS induced depressive-like behaviors and cognitive impairment, high TNF-α and IL-1ß levels, decreased GFAP, and increased Iba-1 expressions. RIP I and RIP II reversed these alterations. These results contribute to the understanding the mechanisms underlying the antidepressant effect of RIP I and RIP II, which may be related to neuroinflammatory suppression.


Asunto(s)
Antidepresivos , Astrocitos , Depresión , Modelos Animales de Enfermedad , Hipocampo , Microglía , Enfermedades Neuroinflamatorias , Estrés Psicológico , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Ratones , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Depresión/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/complicaciones , Estrés Psicológico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Fluoxetina/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-1beta/metabolismo , Fármacos Neuroprotectores/farmacología , Conducta Animal/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo
4.
Behav Brain Res ; 472: 115135, 2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-38964616

RESUMEN

The present study aimed to evaluate the protective potential of carvacrol against depressive-like behavior and cognitive impairment prompted by chronic unpredictable mild stress (CUMS) in mice. The animals were divided into six groups: Control (non-stressed), CARV (carvacrol at 50 mg/kg, p.o.), FLU (fluoxetine at 10 mg/kg, p.o.), CUMS (stressed), CUMS + CARV and CUMS + FLU, and the groups with CUMS were subjected to different stressors for 28 days. After treatment, mice underwent behavioral testing (open field, forced swimming, sucrose preference, social interaction, novel object recognition and Y-maze) and brain areas were removed for oxidative stress (MDA, nitrite/nitrate and GSH levels) and cytokine (IL-1ß and TNF-α) content assays. The results revealed that CARV administration reversed depressive-like behavior and significantly ameliorated the cognitive deficit induced by CUMS, as well as was able to attenuate oxidative stress (decreased MDA and nitrite/nitrate levels and increased GSH levels). In addition, a significant reduction in hippocampal IL-1ß and TNF-α levels was observed, demonstrating a potential anti-neuroinflammatory activity. Taken together, the antioxidant and anti-inflammatory activities observed in this study indicate that CARV is a promising drug for antidepressant treatment.


Asunto(s)
Conducta Animal , Disfunción Cognitiva , Cimenos , Depresión , Modelos Animales de Enfermedad , Enfermedades Neuroinflamatorias , Estrés Oxidativo , Estrés Psicológico , Animales , Estrés Oxidativo/efectos de los fármacos , Ratones , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Disfunción Cognitiva/metabolismo , Depresión/tratamiento farmacológico , Depresión/metabolismo , Depresión/etiología , Masculino , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/complicaciones , Estrés Psicológico/metabolismo , Cimenos/farmacología , Cimenos/administración & dosificación , Conducta Animal/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Antidepresivos/farmacología , Antidepresivos/administración & dosificación , Antioxidantes/farmacología , Fluoxetina/farmacología , Fluoxetina/administración & dosificación , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Interleucina-1beta/metabolismo
5.
J Neurosci ; 44(32)2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38977301

RESUMEN

Overexpression of the agouti-signaling protein (asip1), an endogenous melanocortin antagonist, under the control of a constitutive promoter in zebrafish [Tg(Xla.Eef1a1:Cau.Asip1]iim4] (asip1-Tg) increases food intake by reducing sensitivity of the central satiety systems and abolish circadian activity rhythms. The phenotype also shows increased linear growth and body weight, yet no enhanced aggressiveness in dyadic fights is observed. In fact, asip1-Tg animals choose to flee to safer areas rather than face a potential threat, thus suggesting a potential anxiety-like behavior (ALB). Standard behavioral tests, i.e., the open field test (OFT), the novel object test (NOT), and the novel tank dive test (NTDT), were used to investigate thigmotaxis and ALB in male and female zebrafish. Results showed that the asip1-Tg strain exhibited severe ALB in every test, mainly characterized by pronounced freezing behavior and increased linear and angular swimming velocities. asip1-Tg animals exhibited low central serotonin (5-HT) and dopamine (DA) levels and high turnover rates, thus suggesting that central monoaminergic pathways might mediate melanocortin antagonist-induced ALB. Accordingly, the treatment of asip1-Tg animals with fluoxetine, a selective serotonin reuptake inhibitor (SSRI), reversed the ALB phenotype in NTDT as well as 5-HT turnover. Genomic and anatomical data further supported neuronal interaction between melanocortinergic and serotonergic systems. These results suggest that inhibition of the melanocortin system by ubiquitous overexpression of endogenous antagonist has an anxiogenic effect mediated by serotonergic transmission.


Asunto(s)
Ansiedad , Serotonina , Pez Cebra , Animales , Ansiedad/metabolismo , Ansiedad/psicología , Masculino , Femenino , Serotonina/metabolismo , Animales Modificados Genéticamente , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Fluoxetina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Dopamina/metabolismo , Proteína Relacionada con Agouti/metabolismo , Proteína Relacionada con Agouti/genética
6.
Neurosci Bull ; 40(8): 1037-1052, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39014176

RESUMEN

Posttraumatic stress disorder (PTSD) is a complex mental disorder notable for traumatic experience memory. Although current first-line treatments are linked with clinically important symptom reduction, a large proportion of patients retained to experience considerable residual symptoms, indicating pathogenic mechanism should be illustrated further. Recent studies reported that newly formed myelin could shape neural circuit function and be implicated in fear memory preservation. However, its role in PTSD remains to be elucidated. In this study, we adopted a restraint stress-induced PTSD mouse model and found that PTSD-related neuropsychiatric symptoms were accompanied by increased myelination in the posterior parietal cortex and hippocampus. Fluoxetine, but not risperidone or sertraline, has a more profound rescue effect on neuropsychological behaviors and myelin abnormalities. Further mechanistic experiments revealed that fluoxetine could directly interfere with oligodendroglial differentiation by upregulating Wnt signaling. Our data demonstrated the correlation between PTSD and abnormal myelination, suggesting that the oligodendroglial lineage could be a target for PTSD treatment.


Asunto(s)
Modelos Animales de Enfermedad , Fluoxetina , Ratones Endogámicos C57BL , Vaina de Mielina , Trastornos por Estrés Postraumático , Animales , Trastornos por Estrés Postraumático/tratamiento farmacológico , Fluoxetina/farmacología , Fluoxetina/uso terapéutico , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Vaina de Mielina/metabolismo , Masculino , Ratones , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Lóbulo Parietal/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
7.
Behav Pharmacol ; 35(6): 338-350, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39051900

RESUMEN

Increasing evidence indicates that neuroinflammation, oxidative stress, and neurotrophic factors play a key role in the pathophysiology of major depressive disorder (MDD). In addition, the attenuation of inflammatory response has been considered a putative mechanism for MDD treatment. PT-31 is an imidazolidine derivative and a putative α2-adrenoceptor agonist that has previously demonstrated antinociceptive activity. The present study aimed to investigate the effect of PT-31 on depressive-like behavior and lipopolysaccharide-induced neurochemical changes. To this end, mice received intraperitoneally saline or lipopolysaccharide (600 µg/kg), and 5 h postinjection animals were orally treated with saline, PT-31 (3, 10, and 30 mg/kg), or fluoxetine (30 mg/kg). Mice were subjected to the open field test (OFT) 6 and 24 h after lipopolysaccharide administration and to the tail suspension test (TST) 24 h postlipopolysaccharide. Subsequently, animals were euthanized, and brains were dissected for neurochemical analyses. The administration of lipopolysaccharide-induced sickness- and depressive-like behaviors, besides promoting an increase in myeloperoxidase activity and a reduction in brain-derived neurotrophic factor (BDNF) levels. Noteworthy, PT-31 3 mg/kg attenuated lipopolysaccharide-induced decreased locomotor activity 6 h after lipopolysaccharide in the OFT. All tested doses of PT-31 significantly reduced the immobility time of animals in the TST and attenuated lipopolysaccharide-induced increased myeloperoxidase activity in the cortex of mice. Our results demonstrate that PT-31 ameliorates behavioral changes promoted by lipopolysaccharide in OFT and TST, which is possibly mediated by attenuation of the inflammatory response.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2 , Antidepresivos , Conducta Animal , Factor Neurotrófico Derivado del Encéfalo , Depresión , Lipopolisacáridos , Animales , Lipopolisacáridos/farmacología , Ratones , Masculino , Antidepresivos/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Depresión/tratamiento farmacológico , Depresión/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Fluoxetina/farmacología , Relación Dosis-Respuesta a Droga , Prueba de Campo Abierto/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Suspensión Trasera , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/metabolismo
8.
Nat Commun ; 15(1): 6457, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39085209

RESUMEN

Serotonin reuptake inhibitor antidepressants such as fluoxetine are widely used to treat mood disorders. The mechanisms of action include an increase in extracellular level of serotonin, neurogenesis, and growth of vessels in the brain. We investigated whether fluoxetine could have broader peripheral regenerative properties. Following prolonged administration of fluoxetine in male mice, we showed that fluoxetine increases the number of muscle stem cells and muscle angiogenesis, associated with positive changes in skeletal muscle function. Fluoxetine also improved skeletal muscle regeneration after single and multiples injuries with an increased muscle stem cells pool and vessel density associated with reduced fibrotic lesions and inflammation. Mice devoid of peripheral serotonin treated with fluoxetine did not exhibit beneficial effects during muscle regeneration. Specifically, pharmacological, and genetic inactivation of the 5-HT1B subtype serotonin receptor also abolished the enhanced regenerative process induced by fluoxetine. We highlight here a regenerative property of serotonin on skeletal muscle.


Asunto(s)
Fluoxetina , Músculo Esquelético , Regeneración , Inhibidores Selectivos de la Recaptación de Serotonina , Serotonina , Animales , Masculino , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Regeneración/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Fluoxetina/farmacología , Ratones , Serotonina/metabolismo , Ratones Endogámicos C57BL , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células Madre/citología , Neovascularización Fisiológica/efectos de los fármacos
9.
Sci Rep ; 14(1): 13762, 2024 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-38877103

RESUMEN

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.


Asunto(s)
Fluoxetina , Microbioma Gastrointestinal , Lactancia , ARN Ribosómico 16S , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Femenino , Embarazo , Lactancia/efectos de los fármacos , Fluoxetina/farmacología , Fluoxetina/efectos adversos , Ratones , ARN Ribosómico 16S/genética , Efectos Tardíos de la Exposición Prenatal/microbiología , Inhibidores Selectivos de la Recaptación de Serotonina/efectos adversos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Heces/microbiología , Exposición Materna/efectos adversos , Bacterias/efectos de los fármacos , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación
10.
Eur J Pharmacol ; 978: 176801, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-38945285

RESUMEN

Depression is a serious medical illness characterized by persistent feelings of sadness, hopelessness, and lack of interest in daily activities. It can interfere with daily functioning and quality of life. Despite decades of research, the pathophysiology of depression remains incompletely understood. The correlation between depression and inflammation has recently attracted considerable attention. This study investigated the potential antidepressant effect of etanercept, a tumor necrosis factor-alpha (TNF-α) inhibitor, utilizing a chronic mild stress (CMS) model in rats. Male Wistar rats were divided into two groups; one following a non-stressed protocol and the other a stressed protocol for 5 weeks. From the beginning of the third week, rats were treated either with saline daily or with etanercept twice a week (0.3 mg/kg, i.p.) or with fluoxetine daily (10 mg/kg, i.p) as a reference. Etanercept exhibited comparable effects to those of fluoxetine in counteracting CMS-induced behavioral manifestation in the forced swimming and splash tests. Etanercept also restored serotonin and norepinephrine levels to control values in the prefrontal cortex (PFC). Moreover, the current study verified the antioxidant and anti-inflammatory effects of etanercept. Interestingly, etanercept halted the expression of both norepinephrine and serotonin transporters in stressed rats. This could be attributed to abrogation of the p38 mitogen-activated protein kinase (p38 MAPK) and signal transducer and activator of transcription 3 (STAT-3) pathways in the PFC. The findings of the present study contribute to the understanding of the potential of etanercept as an antidepressant and provide insights into the neurobiological mechanisms underlying its therapeutic effects.


Asunto(s)
Antidepresivos , Conducta Animal , Depresión , Etanercept , Ratas Wistar , Factor de Transcripción STAT3 , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Estrés Psicológico , Animales , Etanercept/farmacología , Etanercept/uso terapéutico , Masculino , Depresión/tratamiento farmacológico , Depresión/metabolismo , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología , Ratas , Factor de Transcripción STAT3/metabolismo , Conducta Animal/efectos de los fármacos , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Norepinefrina/metabolismo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Fluoxetina/farmacología , Fluoxetina/uso terapéutico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Serotonina/metabolismo , Enfermedad Crónica , Transducción de Señal/efectos de los fármacos
11.
Eur J Neurosci ; 60(4): 4491-4502, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38932560

RESUMEN

D-limonene is a widely used flavouring additive in foods, beverages and fragrances due to its pleasant lemon-like odour. This study aimed to investigate the effects of D-limonene on the central nervous system when subjected to chronic restraint stress in rats for 21 days. Forty rats were randomly divided into five groups: i) control, ii) D-limonene, iii) restraint stress, iv) restraint stress+D-limonene and v) restraint stress+fluoxetine. Following the induction of restraint stress, the sucrose preference test, the open field test, the novel object recognition test and the forced swimming test were performed. The levels of BDNF, IL-1ß, IL-6 and caspase-1 were measured from hippocampal tissue using the ELISA method. Sucrose preference test results showed an increase in consumption rate in the stress+D-limonene and a decrease in the stress group. The stress+D-limonene group reversed the increased defensive behaviour observed in the open-field test compared to the stress group. In the novel object recognition test, the discrimination index of the stress+D-limonene group increased compared to the stress group. BDNF levels increased in the stress+limonene group compared to the stress group. In contrast, IL-1ß and caspase-1 levels increased in the stress group compared to the control and decreased in the stress+limonene group compared to the stress group. In this study, D-limonene has been found to have antidepressant-like properties, reducing anhedonic and defensive behaviours and the impairing effects of stress on learning and memory tests. It was observed that D-limonene showed these effects by alleviating neuroinflammation induced by chronic restraint stress in rats.


Asunto(s)
Depresión , Limoneno , Restricción Física , Estrés Psicológico , Animales , Masculino , Limoneno/farmacología , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Ratas , Depresión/tratamiento farmacológico , Memoria/efectos de los fármacos , Ratas Wistar , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Aprendizaje/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Fluoxetina/farmacología , Conducta Animal/efectos de los fármacos , Terpenos/farmacología , Antidepresivos/farmacología
12.
Int J Mol Sci ; 25(12)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38928248

RESUMEN

Fluoxetine, a commonly prescribed medication for depression, has been studied in Alzheimer's disease (AD) patients for its effectiveness on cognitive symptoms. The aim of this systematic review is to investigate the therapeutic potential of fluoxetine in cognitive decline in AD, focusing on its anti-degenerative mechanisms of action and clinical implications. According to PRISMA, we searched MEDLINE, up to 1 April 2024, for animal and human studies examining the efficacy of fluoxetine with regard to the recovery of cognitive function in AD. Methodological quality was evaluated using the ARRIVE tool for animal AD studies and the Cochrane tool for clinical trials. In total, 22 studies were analyzed (19 animal AD studies and 3 clinical studies). Fluoxetine promoted neurogenesis and enhanced synaptic plasticity in preclinical models of AD, through a decrease in Aß pathology and increase in BDNF, by activating diverse pathways (such as the DAF-16-mediated, TGF-beta1, ILK-AKT-GSK3beta, and CREB/p-CREB/BDNF). In addition, fluoxetine has anti-inflammatory properties/antioxidant effects via targeting antioxidant Nrf2/HO-1 and hindering TLR4/NLRP3 inflammasome. Only three clinical studies showed that fluoxetine ameliorated the cognitive performance of people with AD; however, several methodological issues limited the generalizability of these results. Overall, the high-quality preclinical evidence suggests that fluoxetine may have neuroprotective, antioxidant, and anti-inflammatory effects in AD animal models. While more high-quality clinical research is needed to fully understand the mechanisms underlying these effects, fluoxetine is a promising potential treatment for AD patients. If future clinical trials confirm its anti-degenerative and neuroprotective effects, fluoxetine could offer a new therapeutic approach for slowing down the progression of AD.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Fluoxetina , Fluoxetina/uso terapéutico , Fluoxetina/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Humanos , Animales , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Neurogénesis/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos
13.
Eur J Pharmacol ; 976: 176693, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38834095

RESUMEN

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.


Asunto(s)
Antidepresivos , Astrocitos , Depresión , Modelos Animales de Enfermedad , Fluoxetina , Piroptosis , Estrés Psicológico , Arrestina beta 2 , Animales , Fluoxetina/farmacología , Fluoxetina/uso terapéutico , Piroptosis/efectos de los fármacos , Arrestina beta 2/metabolismo , Ratones , Depresión/tratamiento farmacológico , Depresión/metabolismo , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Masculino , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Ratones Endogámicos C57BL , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones Noqueados , Conducta Animal/efectos de los fármacos , Inflamasomas/metabolismo , Enfermedad Crónica
14.
Synapse ; 78(4): e22304, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38896000

RESUMEN

The goal of this report is to explore how K2P channels modulate axonal excitability by using the crayfish ventral superficial flexor preparation. This preparation allows for simultaneous recording of motor nerve extracellular action potentials (eAP) and intracellular excitatory junctional potential (EJP) from a muscle fiber. Previous pharmacological studies have demonstrated the presence of K2P-like channels in crayfish. Fluoxetine (50 µM) was used to block K2P channels in this study. The blocker caused a gradual decline, and eventually complete block, of motor axon action potentials. At an intermediate stage of the block, when the peak-to-peak amplitude of eAP decreased to ∼60%-80% of the control value, the amplitude of the initial positive component of eAP declined at a faster rate than that of the negative peak representing sodium influx. Furthermore, the second positive peak following this sodium influx, which corresponds to the after-hyperpolarizing phase of intracellularly recorded action potentials (iAP), became larger during the intermediate stage of eAP block. Finally, EJP recorded simultaneously with eAP showed no change in amplitude, but did show a significant increase in synaptic delay. These changes in eAP shape and EJP delay are interpreted as the consequence of depolarized resting membrane potential after K2P channel block. In addition to providing insights to possible functions of K2P channels in unmyelinated axons, results presented here also serve as an example of how changes in eAP shape contain information that can be used to infer alterations in intracellular events. This type of eAP-iAP cross-inference is valuable for gaining mechanistic insights here and may also be applicable to other model systems.


Asunto(s)
Potenciales de Acción , Astacoidea , Axones , Fluoxetina , Neuronas Motoras , Animales , Astacoidea/efectos de los fármacos , Astacoidea/fisiología , Fluoxetina/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/fisiología , Axones/efectos de los fármacos , Axones/fisiología
15.
Int J Mol Sci ; 25(12)2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38928021

RESUMEN

Drug repurposing, rebranding an existing drug for a new therapeutic indication, is deemed a beneficial approach for a quick and cost-effective drug discovery process by skipping preclinical, Phase 1 trials and pharmacokinetic studies. Several psychotropic drugs, including selective serotonin reuptake inhibitors (SSRIs) and tricyclic antidepressants (TCAs), were studied for their potential application in different diseases, especially in cancer therapy. Fluoxetine (FLX) is one of the most prescribed psychotropic agents from the SSRIs class for the treatment of several neuropsychiatric disorders with a favorable safety profile. FLX exhibited different oncolytic effects via mechanisms distinct from its main serotonergic activity. Taking advantage of its ability to rapidly penetrate the blood-brain barrier, FLX could be particularly useful in brain tumors. This was proved by different in vitro and in vivo experiments using FLX as a monotherapy or combination with temozolomide (TMZ) or radiotherapy. In this review of the literature, we summarize the potential pleiotropic oncolytic roles of FLX against different cancers, highlighting the multifaceted activities of FLX and its ability to interrupt cancer proliferation via several molecular mechanisms and even surmount multidrug resistance (MDR). We elaborated on the successful synergistic combinations such as FXR/temozolomide and FXR/raloxifene for the treatment of glioblastoma and breast cancer, respectively. We showcased beneficial pharmaceutical trials to load FLX onto carriers to enhance its safety and efficacy on cancer cells. This is the first review article extensively summarizing all previous FLX repurposing studies for the management of cancer.


Asunto(s)
Reposicionamiento de Medicamentos , Fluoxetina , Humanos , Reposicionamiento de Medicamentos/métodos , Fluoxetina/uso terapéutico , Fluoxetina/farmacología , Animales , Neoplasias/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Psicotrópicos/uso terapéutico , Psicotrópicos/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
16.
Neurosci Lett ; 834: 137848, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38823510

RESUMEN

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.


Asunto(s)
Fluoxetina , Ketamina , Corteza Prefrontal , Ratas Wistar , Núcleos Septales , Animales , Ketamina/farmacología , Fluoxetina/farmacología , Masculino , Corteza Prefrontal/efectos de los fármacos , Ratas , Núcleos Septales/efectos de los fármacos , Estimulación Eléctrica
17.
Biomed Pharmacother ; 177: 117017, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38917762

RESUMEN

5-HT clearance, commonly mediated by transporters in the uptake-1 and uptake-2 families, has been linked to 5-HT1B receptor's action on behaviors. Since no specific transporters identified yet, effects of serotonin transporter (SERT) and organic cation transporter (OCTs) on 5-HT1B-elicited immobility phenotype, and 5-HT and HIS uptake were then investigated. Intraperitoneal injections of SERT inhibitor fluoxetine (FLX) and/or OCTs inhibitor decynium (D22) were used prior to local perfusion of 5-HT1B agonist CP93129 into the ventral hippocampus to measure immobility times in the FST and TST, to measure 5-HT uptake efficiencies and HIS uptake efficiencies derived from linear regressions using the transient no-net-flux quantitative microdialysis in C57BL/6 mice. Exogenous 5-HT and HIS uptake were measured following incubation of FLX and/or D22 with CP93129 in the RBL-2H3 cells. Moreover, surface membrane levels of SERT and OCT were detected in response to CP93129. Local CP93129 prolonged immobility times, which were attenuated following pretreatment of either inhibitor. Local CP93129 lowered the slopes obtained from the lineal regressions for 5-HT and HIS (slope is reciprocal to uptake efficiency), which were then weakened following pretreatment of either inhibitor. Similar findings were obtained following CP93129 incubation, and co-incubation of CP93129 with either inhibitor in the RBL-2H3. Moreover, CP93129 dose-dependently moved SERT and OCT3 in the cytosol to the surface membrane. Both SERT and OCT are the target effectors mediating 5-HT1B regulation of immobility time and 5-HT uptake, OCT mediates 5-HT1B regulation of HIS uptake. Their underlying signal transductions need to be further explored.


Asunto(s)
Ratones Endogámicos C57BL , Receptor de Serotonina 5-HT1B , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Serotonina , Animales , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Receptor de Serotonina 5-HT1B/metabolismo , Masculino , Ratones , Conducta Animal/efectos de los fármacos , Fluoxetina/farmacología , Proteínas de Transporte de Catión Orgánico/metabolismo , Ratas , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos
18.
Biol Pharm Bull ; 47(5): 946-954, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38735732

RESUMEN

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.


Asunto(s)
Antiinflamatorios , Fluoxetina , Interleucina-6 , Relación Estructura-Actividad , Animales , Fluoxetina/farmacología , Ratones , Interleucina-6/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/química , Línea Celular , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Citocinas/metabolismo , Receptor Toll-Like 3/metabolismo , Poli I-C/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/química , Inflamación/tratamiento farmacológico
19.
Dev Psychobiol ; 66(5): e22501, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38807259

RESUMEN

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.


Asunto(s)
Miedo , Fluoxetina , Memoria , Corteza Prefrontal , Inhibidores Selectivos de la Recaptación de Serotonina , Fluoxetina/farmacología , Fluoxetina/administración & dosificación , Animales , Miedo/efectos de los fármacos , Miedo/fisiología , Masculino , Ratas , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Corteza Prefrontal/efectos de los fármacos , Memoria/efectos de los fármacos , Memoria/fisiología , Factores de Edad , Ratas Sprague-Dawley , Parvalbúminas/metabolismo , Complejo Nuclear Basolateral/efectos de los fármacos , Complejo Nuclear Basolateral/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Red Nerviosa/efectos de los fármacos
20.
Int Immunopharmacol ; 136: 112335, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38815349

RESUMEN

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.


Asunto(s)
Cloropirifos , Fluoxetina , Janus Quinasa 1 , FN-kappa B , Fármacos Neuroprotectores , PPAR gamma , Factor de Transcripción STAT3 , Transducción de Señal , Sirtuina 1 , Animales , Factor de Transcripción STAT3/metabolismo , Sirtuina 1/metabolismo , FN-kappa B/metabolismo , PPAR gamma/metabolismo , Janus Quinasa 1/metabolismo , Masculino , Fluoxetina/farmacología , Fluoxetina/uso terapéutico , Transducción de Señal/efectos de los fármacos , Cloropirifos/toxicidad , Ratas , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Neuronas/efectos de los fármacos , Neuronas/patología , Estrés Oxidativo/efectos de los fármacos , Insecticidas/toxicidad , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Ratas Sprague-Dawley , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...