Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 415
Filtrar
1.
CNS Neurosci Ther ; 30(5): e14719, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38783536

RESUMEN

BACKGROUND: Methamphetamine (METH) is a psychostimulant substance with highly addictive and neurotoxic effects, but no ideal treatment option exists to improve METH-induced neurocognitive deficits. Recently, mesenchymal stem cells (MSCs)-derived exosomes have raised many hopes for treating neurodegenerative sequela of brain disorders. This study aimed to determine the therapeutic potential of MSCs-derived exosomes on cognitive function and neurogenesis of METH-addicted rodents. METHODS: Male BALB/c mice were subjected to chronic METH addiction, followed by intravenous administration of bone marrow MSCs-derived exosomes. Then, the spatial memory and recognition memory of animals were assessed by the Barnes maze and the novel object recognition test (NORT). The neurogenesis-related factors, including NeuN and DCX, and the expression of Iba-1, a microglial activation marker, were assessed in the hippocampus by immunofluorescence staining. Also, the expression of inflammatory cytokines, including TNF-α and NF-κB, were evaluated by western blotting. RESULTS: The results showed that BMSCs-exosomes improved the time spent in the target quadrant and correct-to-wrong relative time in the Barnes maze. Also, NORT's discrimination index (DI) and recognition index (RI) were improved following exosome therapy. Additionally, exosome therapy significantly increased the expression of NeuN and DCX in the hippocampus while decreasing the expression of inflammatory cytokines, including TNF-α and NF-κB. Besides, BMSC-exosomes down-regulated the expression of Iba-1. CONCLUSION: Our findings indicate that BMSC-exosomes mitigated METH-caused cognitive dysfunction by improving neurogenesis and inhibiting neuroinflammation in the hippocampus.


Asunto(s)
Trastornos Relacionados con Anfetaminas , Proteína Doblecortina , Exosomas , Hipocampo , Células Madre Mesenquimatosas , Metanfetamina , Ratones Endogámicos BALB C , Neurogénesis , Animales , Exosomas/metabolismo , Masculino , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Ratones , Metanfetamina/toxicidad , Trastornos Relacionados con Anfetaminas/terapia , Trastornos Relacionados con Anfetaminas/psicología , Trastornos Relacionados con Anfetaminas/metabolismo , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Cognición/efectos de los fármacos , Cognición/fisiología , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Proteínas del Tejido Nervioso/metabolismo , Estimulantes del Sistema Nervioso Central/toxicidad , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología , Proteínas de Microfilamentos/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Proteínas de Unión al Calcio , Proteínas de Unión al ADN
2.
Physiol Behav ; 281: 114549, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38604593

RESUMEN

Maternal exposure to elevated levels of steroid hormones during pregnancy is associated with the development of chronic conditions in offspring that manifest in adulthood. However, the effects of progesterone (P4) administration during early pregnancy on fetal development and subsequent offspring behavior remain poorly understood. In this study, we aimed to investigate the effects of P4 treatment during early pregnancy on the transcript abundance in the fetal brain and assess the behavioral consequences in the offspring during adolescence and adulthood. Using RNA-seq analysis, we examined the impact of P4 treatment on the fetal brain transcriptome in a dosage-dependent manner. Our results revealed differential regulation of genes involved in neurotransmitter transport, synaptic transmission, and transcriptional regulation. Specifically, we observed bidirectional regulation of transcription factors (TFs) by P4 at different doses, highlighting the critical role of these TFs in neurodevelopment. To assess behavioral outcomes, we conducted open field and elevated plus maze tests. Offspring treated with low-dose P4 (LP4) displayed increased exploratory behavior during both adolescence and adulthood. In contrast, the high-dose P4 (HP4) group exhibited impaired exploration and heightened anxiety-like behaviors compared to the control mice. Moreover, in a novel object recognition test, HP4-treated offspring demonstrated impaired object recognition memory during both developmental stages. Additionally, both LP4 and HP4 groups showed reduced social interaction in the three-chamber test. These results suggest that prenatal exposure to P4 exerts a notable influence on the expression of genes associated with neurodevelopment and may induce alterations in behavioral characteristics in progeny, highlighting the need to monitor progesterone levels during pregnancy for long-term impacts on fetal brain development and behavior.


Asunto(s)
Conducta Animal , Encéfalo , Conducta Exploratoria , Efectos Tardíos de la Exposición Prenatal , Progesterona , Transcriptoma , Animales , Embarazo , Progesterona/farmacología , Femenino , Efectos Tardíos de la Exposición Prenatal/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/crecimiento & desarrollo , Ratones , Transcriptoma/efectos de los fármacos , Masculino , Conducta Animal/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Ansiedad , Ratones Endogámicos C57BL , Reconocimiento en Psicología/efectos de los fármacos , Progestinas/farmacología
3.
Sci Rep ; 12(1): 249, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34997032

RESUMEN

Older adult patients with sepsis frequently experience cognitive impairment. The roles of brain neutrophil gelatinase-associated lipocalin (NGAL) and iron in older sepsis patients remain unknown. We investigated the effects of lipopolysaccharide-induced sepsis on novel object recognition test, NGAL levels, an inflammatory mediator tumor necrosis factor-α (TNFα) levels, and iron ion levels in the hippocampus and cortex of young and aged rats. The effect of an iron chelator deferoxamine pretreatment on aged sepsis rats was also examined. Young sepsis-survivor rats did not show impaired novel object recognition, TNFα responses, or a Fe2+/Fe3+ imbalance. They showed hippocampal and cortical NGAL level elevations. Aged sepsis-survivor rats displayed a decreased object discrimination index, elevation of NGAL levels and Fe2+/Fe3+ ratio, and no TNFα responses. Pretreatment with deferoxamine prevented the reduction in the object recognition of aged sepsis-survivor rats. The elevation in hippocampal and cortical NGAL levels caused by lipopolysaccharide was not influenced by deferoxamine pretreatment. The lipopolysaccharide-induced Fe2+/Fe3+ ratio elevation was blocked by deferoxamine pretreatment. In conclusion, our findings suggest that iron homeostasis in the cortex and hippocampus contributes to the maintenance of object recognition ability in older sepsis survivors.


Asunto(s)
Conducta Animal , Encéfalo/enzimología , Disfunción Cognitiva/enzimología , Hierro/metabolismo , Lipocalina 2/metabolismo , Reconocimiento en Psicología , Sepsis/enzimología , Factores de Edad , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/prevención & control , Disfunción Cognitiva/psicología , Deferoxamina/farmacología , Modelos Animales de Enfermedad , Homeostasis , Masculino , Prueba de Campo Abierto , Ratas Sprague-Dawley , Reconocimiento en Psicología/efectos de los fármacos , Sepsis/tratamiento farmacológico , Sepsis/fisiopatología , Sepsis/psicología , Sideróforos/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
4.
Brain Res Bull ; 178: 120-130, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34838642

RESUMEN

In addition to motor dysfunction, cognitive impairments have been reported to occur in patients with early-stage Parkinson's disease (PD). In this study, we examined a PD mouse model induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). This treatment led to the degeneration of nigrostriatal dopaminergic neurons in mice, a phenomenon that is consistent with previous studies. Besides, spatial memory and object recognition of MPTP-treated mice were impaired, as denoted by the Morris water maze (MWM) and novel object recognition (NOR) tests, respectively. Moreover, hippocampal synaptic plasticity (long-term potentiation and depotentiation) and the levels of synaptic proteins in hippocampus were decreased after MPTP treatment. We also found that MPTP resulted in the microglial activation and an inflammatory response in the striatum and hippocampus. Mammalian asparagine endopeptidase (AEP), a cysteine lysosomal protease, is involved in the cleavage and activation of Toll-like receptors (TLRs). The deletion of AEP can inhibit TLR4 in a mouse model of Alzheimer's disease, and TLR4 is upregulated in PD, inducing microglial activation and inflammation. We found that AEP deletion provided greater resistance to the toxic effects of MPTP. AEP knockout ameliorated the cognition and the synaptic plasticity defects in the hippocampus. Furthermore, AEP deletion decreased the expression of TLR4 and reduced microglial activation and the levels of several proinflammatory cytokines. Thus, we suggest that AEP plays a role in the inflammation induced by MPTP, and TLR4 might also involve in this process. AEP deletion could be a possible treatment strategy for the cognitive deficits of PD.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Disfunción Cognitiva/inducido químicamente , Cisteína Endopeptidasas/metabolismo , Dopaminérgicos/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Intoxicación por MPTP , Enfermedades Neuroinflamatorias/inducido químicamente , Plasticidad Neuronal/efectos de los fármacos , Receptor Toll-Like 4/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/fisiopatología , Ratones , Ratones Noqueados , Reconocimiento en Psicología/efectos de los fármacos , Aprendizaje Espacial/efectos de los fármacos , Memoria Espacial/efectos de los fármacos
5.
Behav Brain Res ; 419: 113677, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34818568

RESUMEN

In long-term spaceflight, astronauts will face unique cognitive loads and social challenges which will be complicated by communication delays with Earth. It is important to understand the central nervous system (CNS) effects of deep spaceflight and the associated unavoidable exposure to galactic cosmic radiation (GCR). Rodent studies show single- or simple-particle combination exposure alters CNS endpoints, including hippocampal-dependent behavior. An even better Earth-based simulation of GCR is now available, consisting of a 33-beam (33-GCR) exposure. However, the effect of whole-body 33-GCR exposure on rodent behavior is unknown, and no 33-GCR CNS countermeasures have been tested. Here astronaut-age-equivalent (6mo-old) C57BL/6J male mice were exposed to 33-GCR (75cGy, a Mars mission dose). Pre-/during/post-Sham or 33-GCR exposure, mice received a diet containing a 'vehicle' formulation alone or with the antioxidant/anti-inflammatory compound CDDO-EA as a potential countermeasure. Behavioral testing beginning 4mo post-irradiation suggested radiation and diet did not affect measures of exploration/anxiety-like behaviors (open field, elevated plus maze) or recognition of a novel object. However, in 3-Chamber Social Interaction (3-CSI), CDDO-EA/33-GCR mice failed to spend more time exploring a holder containing a novel mouse vs. a novel object (empty holder), suggesting sociability deficits. Also, Vehicle/33-GCR and CDDO-EA/Sham mice failed to discriminate between a novel stranger vs. familiarized stranger mouse, suggesting blunted preference for social novelty. CDDO-EA given pre-/during/post-irradiation did not attenuate the 33-GCR-induced blunting of preference for social novelty. Future elucidation of the mechanisms underlying 33-GCR-induced blunting of preference for social novelty will improve risk analysis for astronauts which may in-turn improve countermeasures.


Asunto(s)
Conducta Animal , Disfunción Cognitiva , Radiación Cósmica/efectos adversos , Ácido Oleanólico/análogos & derivados , Exposición a la Radiación/efectos adversos , Reconocimiento en Psicología , Conducta Social , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/efectos de la radiación , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ácido Oleanólico/farmacología , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/efectos de la radiación
6.
Biomed Pharmacother ; 144: 112369, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34715446

RESUMEN

As an N-methyl-D-aspartate (NMDA) receptor inhibitor, ketamine has become a popular recreational substance and currently is used to address treatment-resistant depression. Since heavy ketamine use is associated with persisting psychosis, cognitive impairments, and neuronal damage, the safety of ketamine treatment for depression should be concerned. The nutrient supplement betaine has been shown to counteract the acute ketamine-induced psychotomimetic effects and cognitive dysfunction through modulating NMDA receptors. This study aimed to determine whether the adjunctive or subsequent betaine treatment would improve the enduring behavioral disturbances and hippocampal synaptic abnormality induced by repeated ketamine exposure. Mice received ketamine twice daily for 14 days, either combined with betaine co-treatment or subsequent betaine post-treatment for 7 days. Thereafter, three-chamber social approach test, reciprocal social interaction, novel location/object recognition test, forced swimming test, and head-twitch response induced by serotonergic hallucinogen were monitored. Data showed that the enduring behavioral abnormalities after repeated ketamine exposure, including disrupted social behaviors, recognition memory impairments, and increased depression-like and hallucinogen-induced head-twitch responses, were remarkably improved by betaine co-treatment or post-treatment. Consistently, betaine protected and reversed the reduced hippocampal synaptic activity, such as decreases in field excitatory post-synaptic potentiation (fEPSP), long-term potentiation (LTP), and PSD-95 levels, after repeated ketamine treatment. These results demonstrated that both co-treatment and post-treatment with betaine could effectively prevent and reverse the adverse behavioral manifestations and hippocampal synaptic plasticity after repeated ketamine use, suggesting that betaine can be used as a novel adjunct therapy with ketamine for treatment-resistant depression and provide benefits for ketamine use disorders.


Asunto(s)
Antipsicóticos/farmacología , Conducta Animal/efectos de los fármacos , Betaína/farmacología , Hipocampo/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Psicosis Inducidas por Sustancias/prevención & control , Transmisión Sináptica/efectos de los fármacos , Animales , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Antagonistas de Aminoácidos Excitadores , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Hipocampo/metabolismo , Hipocampo/fisiopatología , Ketamina , Locomoción/efectos de los fármacos , Masculino , Ratones Endogámicos ICR , Prueba de Campo Abierto/efectos de los fármacos , Psicosis Inducidas por Sustancias/etiología , Psicosis Inducidas por Sustancias/fisiopatología , Psicosis Inducidas por Sustancias/psicología , Reconocimiento en Psicología/efectos de los fármacos , Conducta Social , Natación
7.
PLoS One ; 16(9): e0257986, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34587208

RESUMEN

The first symptoms of schizophrenia (SCHZ) are usually observed during adolescence, a developmental period during which first exposure to psychoactive drugs also occurs. These epidemiological findings point to adolescence as critical for nicotine addiction and SCHZ comorbidity, however it is not clear whether exposure to nicotine during this period has a detrimental impact on the development of SCHZ symptoms since there is a lack of studies that investigate the interactions between these conditions during this period of development. To elucidate the impact of a short course of nicotine exposure across the spectrum of SCHZ-like symptoms, we used a phencyclidine-induced adolescent mice model of SCHZ (2.5mg/Kg, s.c., daily, postnatal day (PN) 38-PN52; 10mg/Kg on PN53), combined with an established model of nicotine minipump infusions (24mg/Kg/day, PN37-44). Behavioral assessment began 4 days after the end of nicotine exposure (PN48) using the following tests: open field to assess the hyperlocomotion phenotype; novel object recognition, a declarative memory task; three-chamber sociability, to verify social interaction and prepulse inhibition, a measure of sensorimotor gating. Phencyclidine exposure evoked deficits in all analyzed behaviors. Nicotine history reduced the magnitude of phencyclidine-evoked hyperlocomotion and impeded the development of locomotor sensitization. It also mitigated the deficient sociability elicited by phencyclidine. In contrast, memory and sensorimotor gating deficits evoked by phencyclidine were neither improved nor worsened by nicotine history. In conclusion, our results show for the first time that nicotine history, restricted to a short period during adolescence, does not worsen SCHZ-like symptoms evoked by a phencyclidine-induced mice model.


Asunto(s)
Conducta Animal/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Nicotina/farmacología , Reconocimiento en Psicología/efectos de los fármacos , Esquizofrenia/tratamiento farmacológico , Filtrado Sensorial/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Femenino , Locomoción/efectos de los fármacos , Masculino , Ratones , Nicotina/uso terapéutico , Fenciclidina , Esquizofrenia/inducido químicamente
8.
Brain Res Bull ; 176: 8-17, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34391822

RESUMEN

Gut microbiota dysbiosis is associated with cognitive dysfunctions and Alzheimer's disease (AD). This study set out to better understand the relationship between gut microbiota depletion and cognitive abilities in mice with or without Alzheimer-like disease. Male C57BL/6 mice from early adolescence received an antibiotic cocktail, and then in adulthood, animals were subjected to a stereotaxic surgery to induce Alzheimer-like disease using amyloid-beta (Aß) 1-42 microinjection. To assess cognitive functions in mice, three behavioural tests including the Y maze, object recognition, and Morris water maze were used. We also measured brain-derived-neurotrophic factor (BDNF), tumour-necrosis factor (TNF)-α, interleukin (IL)-6, and Aß42 in the brain. Our findings showed that antibiotics treatment impaired object recognition memory, whereas did not alter spatial memory in healthy mice. Antibiotics treatment in mice significantly exacerbated spatial memory impairment following the induction of AD in both the Y maze and Morris water maze test. There were significant correlations between these behavioural tests. In addition, healthy animals treated with antibiotics displayed a significant reduction in brain IL-6. We observed that antibiotics treatment significantly decreased both cytokines TNF-α and IL-6 in the brain of AD-induced mice. However, no alterations were found in brain BDNF levels following both antibiotics treatment and AD induction. These findings show that antibiotic-induced gut microbiota depletion from early adolescence to adulthood can impair cognitive abilities in mice with or without Alzheimer-like disease. Overall, this study suggests that gut microbiota manipulation from early adolescence to adulthood may adversely affect the normal development of cognitive functions.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Antibacterianos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Reconocimiento en Psicología/efectos de los fármacos , Memoria Espacial/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Interleucina-6/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Factor de Necrosis Tumoral alfa/metabolismo
9.
Neurobiol Learn Mem ; 184: 107499, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34352396

RESUMEN

Estrogens, particularly 17ß-estradiol (estradiol, E2), regulate memory formation. E2 acts through its intracellular receptors, estrogen receptors (ER) ERα and ERß, as well as a recently identified G protein-coupled estrogen receptor (GPER). Although the effects of E2 on memory have been investigated, studies examining the effects of GPER stimulation are scarce. Selective GPER agonism improves memory in ovariectomized female rats, but little information is available regarding the effects of GPER stimulation in male rodents. The aim of the present study was to investigate the effects of the GPER agonist, G1, on consolidation and reconsolidation of inhibitory avoidance (IA) and object recognition (OR) memory in male rats. Animals received vehicle, G1 (15, 75, 150 µg/kg; i.p.), or the GPER antagonist G15 (100 µg/kg; i.p.) immediately after training, or G1 (150 µg/kg; i.p.) 3 or 6 h after training. To investigate reconsolidation, G1 was administered immediately after IA retention Test 1. Results indicated that G1 administered immediately after training at the highest dose enhanced both OR and IA memory consolidation, while GPER blockade immediately after training impaired OR. No effects of GPER stimulation were observed when G1 was given 3 or 6 h after training or after Test 1. The present findings provide evidence that GPER is involved in the early stages of memory consolidation in both neutral and emotional memory tasks in male adult rats.


Asunto(s)
Memoria/fisiología , Receptores Acoplados a Proteínas G/fisiología , Reconocimiento en Psicología/fisiología , Animales , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Antagonistas del Receptor de Estrógeno/farmacología , Estrógenos/farmacología , Masculino , Memoria/efectos de los fármacos , Motivación/fisiología , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/efectos de los fármacos , Reconocimiento en Psicología/efectos de los fármacos
10.
J Pharmacol Sci ; 147(1): 58-61, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34294373

RESUMEN

Nicotine administration enhances object recognition memory. However, target brain regions and cellular mechanisms underlying the nicotine effects remain unclear. In mice, the novel object recognition test revealed that systemic nicotine administration before training enhanced object recognition memory. Moreover, this effect was inhibited by infusion of retigabine, a selective voltage-dependent potassium 7 (Kv7) channel opener, into the medial prefrontal cortex (mPFC) before nicotine administration. Additionally, infusion of XE-991, a selective Kv7 channel blocker, into the mPFC before training enhanced object recognition memory. Therefore, Kv7 channels in the mPFC may be at least partly involved in nicotine-induced enhancement of object recognition memory.


Asunto(s)
Memoria/efectos de los fármacos , Nicotina/farmacología , Canales de Potasio con Entrada de Voltaje/metabolismo , Corteza Prefrontal/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Animales , Antracenos/farmacología , Carbamatos/farmacología , Masculino , Ratones Endogámicos C57BL , Fenilendiaminas/farmacología , Canales de Potasio con Entrada de Voltaje/fisiología , Estimulación Química
11.
Biol Pharm Bull ; 44(7): 1007-1013, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34193682

RESUMEN

Nicotine has been known to enhance recognition memory in various species. However, the brain region where nicotine acts and exerts its effect remains unclear. Since the medial prefrontal cortex (mPFC) is associated with memory, we examined the role of the mPFC in nicotine-induced enhancement of recognition memory using the novel object recognition test in male C57BL/6J mice. Systemic nicotine administration 10 min before training session significantly enhanced object recognition memory in test session that was performed 24 h after the training. Intra-mPFC infusion of mecamylamine, a non-selective nicotinic acetylcholine receptor (nAChR) antagonist, 5 min before nicotine administration blocked the effect of nicotine. Additionally, intra-mPFC infusion of dihydro-ß-erythroidine, a selective α4ß2 nAChR antagonist, or methyllycaconitine, a selective α7 nAChR antagonist, significantly suppressed the nicotine-induced object recognition memory enhancement. Finally, intra-mPFC infusion of nicotine 1 min before the training session augmented object recognition memory in a dose-dependent manner. These findings suggest that mPFC α4ß2 and α7 nAChRs mediate the nicotine-induced object recognition memory enhancement.


Asunto(s)
Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Corteza Prefrontal/efectos de los fármacos , Receptores Nicotínicos/fisiología , Reconocimiento en Psicología/efectos de los fármacos , Receptor Nicotínico de Acetilcolina alfa 7/fisiología , Aconitina/análogos & derivados , Aconitina/farmacología , Animales , Dihidro-beta-Eritroidina/farmacología , Masculino , Mecamilamina/farmacología , Ratones Endogámicos C57BL , Antagonistas Nicotínicos/farmacología , Corteza Prefrontal/fisiología , Receptor Nicotínico de Acetilcolina alfa 7/antagonistas & inhibidores
12.
Mol Neurobiol ; 58(9): 4251-4267, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33970453

RESUMEN

Neuroinflammation can cause cognitive deficits, and preexisting neuroinflammation is observed frequently in the clinic after trauma, surgery, and infection. Patients with preexisting neuroinflammation often need further medical treatment under general anesthesia. However, the effects of postconditioning with general anesthetics on preexisting neuroinflammation have not been determined. In this study, adult rats were posttreated with sevoflurane or propofol after intracerebroventricular administration of lipopolysaccharide. The effects of sevoflurane or propofol postconditioning on neuroinflammation-induced recognition memory deficits were detected. Our results found that postconditioning with sevoflurane but not propofol reversed the selective spatial recognition memory impairment induced by neuroinflammation, and these differential effects did not appear to be associated with the similar anti-neuroinflammatory responses of general anesthetics. However, postconditioning with propofol induced a selective long-lasting upregulation of extrasynaptic NR2B-containing N-methyl-D-aspartate receptors in the dorsal hippocampus, which downregulated the cAMP response element-binding signaling pathway and impaired spatial recognition memory. Additionally, the NR2B antagonists memantine and Ro25-6981 reversed this neurotoxicity induced by propofol postconditioning. Taken together, these results indicate that under preexisting neuroinflammation, postconditioning with sevoflurane can provide reliable neuroprotection by attenuating lipopolysaccharide-induced neuroinflammation, apoptosis, and neuronal loss and eventually improving spatial recognition deficits. However, although posttreatment with propofol also has the same anti-neuroinflammatory effects, the neurotoxicity caused by propofol postconditioning following neuroinflammation warrants further consideration.


Asunto(s)
Cognición/efectos de los fármacos , Hipocampo/efectos de los fármacos , Enfermedades Neuroinflamatorias/metabolismo , Propofol/administración & dosificación , Receptores de N-Metil-D-Aspartato/metabolismo , Sevoflurano/administración & dosificación , Animales , Hipocampo/metabolismo , Lipopolisacáridos , Masculino , Ratas , Ratas Sprague-Dawley , Reconocimiento en Psicología/efectos de los fármacos
13.
Neurobiol Learn Mem ; 182: 107463, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34015440

RESUMEN

Growing evidences indicate that neuropathic pain is frequently accompanied with cognitive impairments, which aggravate the decrease in the quality of life of chronic pain patients. Furthermore, it has been shown that the activation of Glucagon-like-peptide-1receptor (GLP-1R) improved memory deficit in multiple diseases, including Alzheimer's disease (AD), stroke. However, whether GLP-1R activation could improve memory impairment induced by neuropathic pain and the mechanisms underlying the effect of the activation of GLP-1R on memory protection have not yet been established. The spared nerve injury (SNI) model was established as a kind of neuropathic pain. And novel-object recognition memory (hippocampus-dependent memory) was tested by the novel object recognition test (NORT). The expression levels of GLP-1, GLP-1R, adenosine monophosphate-activated protein kinase (AMPK), p-AMPKThr172, nuclear factor κ B p65 (NF-κB p65), interleukin-1beta (IL-1ß), IL-1ß p17 (mature IL-1ß), tumor necrosis factor-alpha (TNF-α) and the synaptic proteins were tested in the murine hippocampus with memory deficits caused by neuropathic pain. Then, exenatide acetate (Ex-4, a GLP-1R agonist), exendin (9-39) (Ex(9-39), a GLP-1R antagonist) and Compound C dihydrochloride (CC, an AMPK inhibitor) were used to test the effects of the activation of GLP-1R in the mice with neuropathic pain. First, we uncovered that neuropathic pain could inhibit GLP-1/GLP-R axis, disturb inflammatory signaling pathway, increase the expression of IL-1ß, IL-1ß p17 and TNF-α, downregulate the synaptic proteins (postsynaptic density protein 95 (PSD95) and Arc). Subsequently, we reported that Ex-4 treatment could improve recognition memory impairment, increase the ratio of p-AMPKThr172/AMPK, inhibit the phosphorylation NF-κB p65 and decrease the expression of IL-1ß, IL-1ß p17 and TNF-α, upregulate the levels of PSD95 and Arc. Moreover, we found that Ex(9-39) and CC treatment could abrogate the memory protection of activation of GLP-1R in mice with neuropathic pain. The results indicated that the activation of GLP-1R could improve recognition memory impairment via regulating AMPK/NF-κB pathway, improving neuroinflammation, reversing the decreased level of synaptic proteins in neuropathic pain mice.


Asunto(s)
Quinasas de la Proteína-Quinasa Activada por el AMP/efectos de los fármacos , Exenatida/farmacología , Receptor del Péptido 1 Similar al Glucagón/agonistas , Hipocampo/efectos de los fármacos , Neuralgia/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Factor de Transcripción ReIA/efectos de los fármacos , Quinasas de la Proteína-Quinasa Activada por el AMP/metabolismo , Animales , Dolor Crónico/metabolismo , Dolor Crónico/fisiopatología , Modelos Animales de Enfermedad , Péptido 1 Similar al Glucagón/efectos de los fármacos , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Hipocampo/metabolismo , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/metabolismo , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/fisiopatología , Ratones , Neuralgia/fisiopatología , Enfermedades Neuroinflamatorias/metabolismo , Prueba de Campo Abierto , Fragmentos de Péptidos/farmacología , Traumatismos de los Nervios Periféricos , Nervio Ciático/cirugía , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
14.
Mar Drugs ; 19(4)2021 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-33801706

RESUMEN

We recently found that dietary supplementation with the seaweed Sargassum fusiforme, containing the preferential LXRß-agonist 24(S)-saringosterol, prevented memory decline and reduced amyloid-ß (Aß) deposition in an Alzheimer's disease (AD) mouse model without inducing hepatic steatosis. Here, we examined the effects of 24(S)-saringosterol as a food additive on cognition and neuropathology in AD mice. Six-month-old male APPswePS1ΔE9 mice and wildtype C57BL/6J littermates received 24(S)-saringosterol (0.5 mg/25 g body weight/day) (APPswePS1ΔE9 n = 20; C57BL/6J n = 19) or vehicle (APPswePS1ΔE9 n = 17; C57BL/6J n = 19) for 10 weeks. Cognition was assessed using object recognition and object location tasks. Sterols were analyzed by gas chromatography/mass spectrometry, Aß and inflammatory markers by immunohistochemistry, and gene expression by quantitative real-time PCR. Hepatic lipids were quantified after Oil-Red-O staining. Administration of 24(S)-saringosterol prevented cognitive decline in APPswePS1ΔE9 mice without affecting the Aß plaque load. Moreover, 24(S)-saringosterol prevented the increase in the inflammatory marker Iba1 in the cortex of APPswePS1ΔE9 mice (p < 0.001). Furthermore, 24(S)-saringosterol did not affect the expression of lipid metabolism-related LXR-response genes in the hippocampus nor the hepatic neutral lipid content. Thus, administration of 24(S)-saringosterol prevented cognitive decline in APPswePS1ΔE9 mice independent of effects on Aß load and without adverse effects on liver fat content. The anti-inflammatory effects of 24(S)-saringosterol may contribute to the prevention of cognitive decline.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antiinflamatorios/farmacología , Conducta Animal/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Cognición/efectos de los fármacos , Nootrópicos/farmacología , Estigmasterol/análogos & derivados , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Animales , Proteínas de Unión al Calcio/metabolismo , Línea Celular Tumoral , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Estigmasterol/farmacología
15.
Neurobiol Learn Mem ; 180: 107423, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33705861

RESUMEN

Social recognition memory (SRM) forms the basis of social relationships of animals. It is essential for social interaction and adaptive behavior, reproduction and species survival. Evidence demonstrates that social deficits of psychiatric disorders such as autism and schizophrenia are caused by alterations in SRM processing by the hippocampus and amygdala. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) and its receptors PAC1, VPAC1 and VPAC2 are highly expressed in these regions. PACAP is a pleiotropic neuropeptide that modulates synaptic function and plasticity and is thought to be involved in social behavior. PACAP signaling also stimulates the nitric oxide (NO) production and targets outcomes to synapses. In the present work, we investigate the effect of the infusion of PACAP-38 (endogenous neuropeptide and potent stimulator of adenylyl cyclase), PACAP 6-38 (PAC1/VPAC2 receptors antagonist) and S-Nitroso-N-acetyl-DL-penicillamine (SNAP, NO donor) in the CA1 region of the hippocampus and in the basolateral amygdala (BLA) on the consolidation of SRM. For this, male Wistar rats with cannulae implanted in CA1 or in BLA were subjected to a social discrimination paradigm, which is based on the natural ability of rodents to investigate unfamiliar conspecifics more than familiar one. In the sample phase (acquisition), animals were exposed to a juvenile conspecific for 1 h. Immediately, 60 or 150 min after, animals received one of different pharmacological treatments. Twenty-four hours later, they were submitted to a 5 min retention test in the presence of the previously presented juvenile (familiar) and a novel juvenile. Animals that received infusions of PACAP 6-38 (40 pg/side) into CA1 immediately after the sample phase or into BLA immediately or 60 min after the sample phase were unable to recognize the familiar juvenile during the retention test. This impairment was abolished by the coinfusion of PACAP 6-38 plus SNAP (5 µg/side). These results show that the blockade of PACAP/PAC1/VPAC2 signaling in the CA1 and BLA during a restricted post-acquisition time window impairs the consolidation of SRM and that the SNAP is able to abolish this deficit. Findings like this could potentially be used in the future to influence studies of psychiatric disorders involving social behavior.


Asunto(s)
Complejo Nuclear Basolateral/efectos de los fármacos , Región CA1 Hipocampal/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/efectos de los fármacos , Reconocimiento en Psicología/efectos de los fármacos , Percepción Social/efectos de los fármacos , Animales , Complejo Nuclear Basolateral/metabolismo , Región CA1 Hipocampal/metabolismo , Consolidación de la Memoria/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Ratas , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/efectos de los fármacos , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/efectos de los fármacos , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/efectos de los fármacos , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Reconocimiento en Psicología/fisiología , S-Nitroso-N-Acetilpenicilamina/farmacología
16.
Horm Behav ; 127: 104873, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33069752

RESUMEN

Although pathogen threat affects social and sexual responses across species, relatively little is known about the underlying neuroendocrine mechanisms. Progesterone has been speculated to be involved in the mediation of pathogen disgust in women, though with mixed experimental support. Here we considered the effects of acute progesterone on the disgust-like avoidance responses of female mice to pathogen threat. Estrous female mice discriminated and avoided the urinary and associated odors of males subclinically infected with the murine nematode parasite, Heligmosomoides polygyrus. These avoidance responses were not significantly affected by pre-treatment with progesterone. Likewise, brief (1 min) exposure to the odors of infected males attenuated the subsequent responses of females to the odors of the normally preferred unfamiliar males and enhanced their preferences for familiar males. Neither progesterone nor allopregnanolone, a central neurosteroid metabolite of progesterone, had any significant effects on the avoidance of unfamiliar males elicited by pre-exposure to a parasitized male. Progesterone and allopregnanolone, did, however, significantly attenuate the typical preferences of estrous females for unfamiliar uninfected males, suggestive of effects on social recognition. These findings with mice indicate that progesterone may have minimal effects on the responses to specific parasite threat and the expression of pathogen disgust but may influence more general social recognition and preferences.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Nematospiroides dubius , Progesterona/farmacología , Reconocimiento en Psicología/efectos de los fármacos , Infecciones por Strongylida/prevención & control , Animales , Animales no Consanguíneos , Asco , Estro/efectos de los fármacos , Estro/fisiología , Femenino , Masculino , Ratones , Nematospiroides dubius/patogenicidad , Odorantes , Reconocimiento en Psicología/fisiología , Conducta Sexual Animal/efectos de los fármacos , Conducta Sexual Animal/fisiología , Infecciones por Strongylida/transmisión
17.
Biomed Res Int ; 2020: 3193725, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33381547

RESUMEN

Traumatic brain injury (TBI) is among the most debilitating neurological disorders with inadequate therapeutic options. It affects all age groups globally leading to post-TBI behavioral challenges and life-long disabilities requiring interventions for these health issues. In the current study, C57BL/6J mice were induced with TBI through the weight-drop method, and outcomes of acutely administered ketamine alone and in combination with perampanel were observed. The impact of test drugs was evaluated for post-TBI behavioral changes by employing the open field test (OFT), Y-maze test, and novel object recognition test (NOR). After that, isolated plasma and brain homogenates were analyzed for inflammatory modulators, i.e., NF-κB and iNOS, through ELISA. Moreover, metabolomic studies were carried out to further authenticate the TBI rescuing potential of drugs. The animals treated with ketamine-perampanel combination demonstrated improved exploratory behavior in OFT (P < 0.05), while ketamine alone as well as in combination yielded anxiolytic effect (P < 0.05-0.001) in posttraumatic mice. Similarly, the % spontaneous alternation and % discrimination index were increased after the administration of ketamine alone (P < 0.05) and ketamine-perampanel combination (P < 0.01-0.001) in the Y-maze test and NOR test, respectively. ELISA demonstrated the reduced central and peripheral expression of NF-κB (P < 0.05) and iNOS (P < 0.01-0.0001) after ketamine-perampanel polypharmacy. The TBI-imparted alteration in plasma metabolites was restored by drug combination as evidenced by metabolomic studies. The outcomes were fruitful with ketamine, but the combination therapy proved more significant in improving all studied parameters. The benefits of this new investigated polypharmacy might be due to their antiglutamatergic, antioxidant, and neuroprotective capacity.


Asunto(s)
Conducta Animal/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Ketamina/administración & dosificación , Piridonas/administración & dosificación , Animales , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Aprendizaje por Laberinto , Metabolómica , Ratones , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/metabolismo , Neuroprotección , Fármacos Neuroprotectores/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitrilos , Reconocimiento en Psicología/efectos de los fármacos , Memoria Espacial/efectos de los fármacos
18.
Behav Pharmacol ; 31(8): 787-791, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32815898

RESUMEN

The present study evaluated the acute effects of the 5-HT3 receptor antagonist, tropisetron, on recognition memory in ovariectomized adult female rats. The non-spatial novel object recognition task was used to assess recognition memory. In this task, ovariectomized rats explored two identical objects during Trial 1. Immediately after Trial 1, rats were primed either with oil, 250 µg progesterone, 20 µg of estrogen, or 20 µg of estrogen + 250 µg progesterone. Four hours later, the test trial (Trial 2) was initiated. Thirty minutes before Trial 2, rats were injected intraperitoneally with either saline, 1.5 or 2.5 mg/Kg tropisetron. During Trial 2, one arm of the T maze contained an object from Trial 1 (familiar or previously encountered), and a new object (novel) was introduced into the other arm. Exploration times with the novel and familiar objects were recorded and data were converted to percent time spent with the novel object. In oil-primed ovariectomized female rats, treatment with 2.5 mg/Kg tropisetron significantly increased percent time with the novel object. Hormonal-priming with estrogen, progesterone, or estrogen + progesterone did not further accentuate the effects of tropisetron. These results suggest that although tropisetron, estrogen, and progesterone all act as antagonists at the 5-HT3 receptors and blocking 5-HT3 receptors enhances cognition, there appears to be no interaction between tropisetron and these hormones on object recognition.


Asunto(s)
Memoria/fisiología , Reconocimiento en Psicología/efectos de los fármacos , Tropisetrón/farmacología , Animales , Cognición/efectos de los fármacos , Estrógenos/farmacología , Femenino , Indoles/farmacología , Memoria/efectos de los fármacos , Ovariectomía , Progesterona/farmacología , Ratas , Receptores de Serotonina 5-HT3/metabolismo , Reconocimiento en Psicología/fisiología , Antagonistas del Receptor de Serotonina 5-HT3/farmacología , Tropisetrón/metabolismo
19.
Synapse ; 75(2): e22186, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32780904

RESUMEN

Metabolic syndrome (MS) results from excessive consumption of high-calorie foods and sedentary lifestyles. Clinically, insulin resistance, abdominal obesity, hyperglycemia, dyslipidemia, and hypertension are observed. MS has been considered a risk factor in the development of dementia. In the brain, a metabolically impaired environment generates oxidative stress and excessive production of pro-inflammatory cytokines that deteriorate the morphology and neuronal function in the hippocampus, leading to cognitive impairment. Therapeutic alternatives suggest that phenolic compounds can be part of the treatment for neuropathies and metabolic diseases. In recent years, the use of Gallic Acid (GA) has demonstrated antioxidant and anti-inflammatory effects that contribute to neuroprotection and memory improvement in animal models. However, the effect of GA on hippocampal neurodegeneration and memory impairment under MS conditions is still unclear. In this work, we administered GA (20 mg/kg) for 60 days to rats with MS. The results show that GA treatment improved zoometric and biochemical parameters, as well as the recognition memory, in animals with MS. Additionally, GA administration increased hippocampal dendritic spines and decreased oxidative stress and inflammation. Our results show that GA treatment improves metabolism: reducing the oxidative and inflammatory environment that facilitates the recovery of the neuronal morphology in the hippocampus of rats with MS. Consequently, the recognition of objects by these animals, suggesting that GA could be used therapeutically in metabolic disorders that cause dementia.


Asunto(s)
Ácido Gálico/farmacología , Hipocampo/efectos de los fármacos , Síndrome Metabólico/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Catalasa/efectos de los fármacos , Catalasa/metabolismo , Dendritas/efectos de los fármacos , Dendritas/patología , Hipocampo/metabolismo , Hipocampo/patología , Inflamación/metabolismo , Insulina/sangre , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/metabolismo , Memoria/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
20.
Mol Cell Endocrinol ; 518: 110985, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32805333

RESUMEN

INTRODUCTION: The 17ß-estradiol (E2) enhances hippocampal dendritic spine synapses, facilitates learning processes, and exerts neuroprotection. Brain estrogen decline has been reported in Alzheimer's disease. The role of GnRH in modulating steroid biosynthesis convinced us to examine whether hippocampal GnRH administration could enhance the local E2 levels and overcome the development of cognition decline in amyloid ß (Aß) neurotoxicity. To explore if GnRH acts through regulating E2 synthesis, letrozole, an aromatase inhibitor, has been applied in combination with GnRH. METHODS: Female rats received an intracerebroventricular injection of Aß. The GnRH and, or letrozole were injected into the CA1 for 14 consecutive days. Working memory, novel object recognition memory, and anxiety-like behavior were evaluated. Serum and hippocampal E2 levels were measured. Hippocampal mRNA expression of GnRH (GnRH-R) and E2 (ERα and ERß) receptors was assessed. GnRH effect on the excitability of pyramidal cells was studied by in vivo single-unit recording. RESULTS: GnRH increased hippocampal E2 levels, evoked an increase in the spontaneous firing of pyramidal neurons, and caused mRNA overexpression of hippocampal GnRH receptors. GnRH prevented the adverse effects of Aß on working memory, NOR index, and anxiogenic behavior. Letrozole did not reverse GnRH modulatory effects on hippocampal E2 levels and neuroprotection. CONCLUSION: GnRH prevented the Aß-induced memory deficit, which may be mediated through hippocampal E2 levels enhancement. The electrophysiological analysis revealed the enhanced neuronal excitability in the CA1 region. All these data suggest that GnRH might be a promising candidate that reduces anxiety and improves memory indices in the context of Aß neurotoxicity.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Ansiedad/tratamiento farmacológico , Disfunción Cognitiva/prevención & control , Estradiol/metabolismo , Hormona Liberadora de Gonadotropina/administración & dosificación , Animales , Ansiedad/inducido químicamente , Ansiedad/metabolismo , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/psicología , Modelos Animales de Enfermedad , Estradiol/sangre , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Letrozol/administración & dosificación , Letrozol/farmacología , Memoria a Corto Plazo/efectos de los fármacos , Ratas , Receptores LHRH/genética , Receptores LHRH/metabolismo , Reconocimiento en Psicología/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA