Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35055113

RESUMEN

Methionine (Met) is considered the most toxic amino acid in mammals. Here, we investigated biochemical and behavioral impacts of ad libitum one-week feeding of high-Met diets on mice. Adult male mice were fed the standard rodent diet that contained 0.44% Met (1×) or a diet containing 16 graded Met doses (1.2×-13×). High-Met diets for one-week induced a dose-dependent decrease in body weight and an increase in serum Met levels with a 2.55 mM peak (versus basal 53 µM) on the 12×Met diet. Total homocysteine (Hcy) levels were also upregulated while concentrations of other amino acids were almost maintained in serum. Similarly, levels of Met and Hcy (but not the other amino acids) were highly elevated in the cerebrospinal fluids of mice on the 10×Met diet; the Met levels were much higher than Hcy and the others. In a series of behavioral tests, mice on the 10×Met diet displayed increased anxiety and decreased traveled distances in an open-field test, increased activity to escape from water soaking and tail hanging, and normal learning/memory activity in a Y-maze test, which were reflections of negative/positive symptoms and normal cognitive function, respectively. These results indicate that high-Met ad libitum feeding even for a week can induce bipolar disorder-like disease models in mice.


Asunto(s)
Trastorno Bipolar/psicología , Homocisteína/sangre , Metionina/efectos adversos , Aminoácidos/sangre , Aminoácidos/líquido cefalorraquídeo , Animales , Trastorno Bipolar/sangre , Trastorno Bipolar/líquido cefalorraquídeo , Trastorno Bipolar/inducido químicamente , Modelos Animales de Enfermedad , Esquema de Medicación , Homocisteína/líquido cefalorraquídeo , Masculino , Metionina/sangre , Metionina/líquido cefalorraquídeo , Ratones , Prueba de Campo Abierto/efectos de los fármacos , Regulación hacia Arriba
2.
Behav Brain Res ; 417: 113615, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-34606775

RESUMEN

The increasing use of aluminum nanoparticles (nano-Al) leads to increased human exposure and might affect human health. Considering the suggested connection between aluminum exposure and Alzheimer's disease (AD) pathogenesis, there is a concern about the effect of nano-Al on cognitive function and brain health. This study was aimed to assess the effect of a 5-day oral gavage of aluminum oxide nanoparticle (nano-Al) on memory and the phosphorylation levels of hippocampal p38, JNK (c-Jun N-terminal kinase), ERK (extracellular signal-regulated kinase) as well as cleaved caspase-3 in mice. Adult male NMRI mice were treated with nano-Al in doses 5 and 10 mg/kg/oral gavage for 5 days. The test session of novel object recognition (NOR) task was performed on day 5. Following the NOR test, the hippocampi were isolated for western blot analysis to determine the total and phosphorylated levels of p38, JNK, ERK as well as cleaved caspase-3 proteins. The results showed that nano-Al oral gavage in doses of 5 and 10 mg/kg impairs NOR memory in mice. Moreover, the memory impairing effect of nano-Al coincided with a dose dependent increase in phosphorylated p38 and cleaved caspase-3 in the hippocampus. It also increased the ratio of phosphorylated to total content of ERK in the hippocampus while JNK signaling was not affected by nano-Al. This study showed that nano-Al in doses as low as 5 and 10 mg/ kg ingested for 5 days impairs NOR memory and activates p38, ERK and cleaved caspase-3 in the hippocampus.


Asunto(s)
Aluminio/administración & dosificación , Caspasa 3/metabolismo , Relación Dosis-Respuesta a Droga , Hipocampo/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Nanopartículas , Prueba de Campo Abierto/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Memoria/efectos de los fármacos , Ratones , Percepción Visual
3.
Chem Biol Interact ; 351: 109767, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34863679

RESUMEN

The present study investigated the role of neuroinflammation and brain oxidative stress induced by neonatal treatment with lipopolysaccharides (LPS) on the development of autism spectrum disorder (ASD)-like behaviors and disruptive hippocampal neurogenesis in rats by exploring the chemopreventive effects of alpha-glycosyl isoquercitrin (AGIQ) as an antioxidant. AGIQ was dietary administered to dams at 0.25% or 0.5% (w/w) from gestational day 18 until postnatal day (PND) 21 on weaning and then to pups until the adult stage on PND 77. The pups were intraperitoneally injected with LPS (1 mg/kg body weight) on PND 3. At PND 6, LPS alone increased Iba1+ and CD68+ cell numbers without changing the CD163+ cell number and strongly upregulated pro-inflammatory cytokine gene expression (Il1a, Il1b, Il6, Nfkb1, and Tnf) in the hippocampus, and increased brain malondialdehyde levels. At PND 10, pups decreased ultrasonic vocalization (USV), suggesting the induction of pro-inflammatory responses and oxidative stress to trigger communicative deficits. By contrast, LPS alone upregulated Nfe2l2 expression at PND 6, increased Iba1+, CD68+, and CD163+ cell numbers, and upregulated Tgfb1 at PND 21, suggesting anti-inflammatory responses until the weaning period. However, LPS alone disrupted hippocampal neurogenesis at weaning and suppressed social interaction parameters and rate of freezing time at fear acquisition and extinction during the adolescent stage. On PND 77, neuroinflammatory responses had mostly disappeared; however, disruptive neurogenesis and fear memory deficits were sustained. AGIQ ameliorated most changes on acute pro-inflammatory responses and oxidative stress at PND 6, and the effects on USVs at PND 10 and neurogenesis and behavioral parameters throughout the adult stage. These results suggested that neonatal LPS treatment induced acute but transient neuroinflammation, triggering the progressive disruption of hippocampal neurogenesis leading to abnormal behaviors in later life. AGIQ treatment was effective for ameliorating LPS-induced progressive changes by critically suppressing initial pro-inflammatory responses and oxidative stress.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Trastorno Autístico/tratamiento farmacológico , Glicósidos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Quercetina/análogos & derivados , Animales , Animales Recién Nacidos , Trastorno Autístico/inducido químicamente , Trastorno Autístico/patología , Giro Dentado/efectos de los fármacos , Giro Dentado/patología , Femenino , Expresión Génica/efectos de los fármacos , Lipopolisacáridos , Masculino , Neurogénesis/efectos de los fármacos , Enfermedades Neuroinflamatorias/inducido químicamente , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/patología , Prueba de Campo Abierto/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Embarazo , Quercetina/uso terapéutico , Ratas Sprague-Dawley , Interacción Social/efectos de los fármacos
4.
Nitric Oxide ; 119: 29-40, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34896554

RESUMEN

Nitric oxide is a small gaseous molecule that plays important roles in the majority of biological functions. Impairments of NO-related pathways contribute to the majority of neurological disorders, such as Alzheimer's disease (AD), and mental disorders, such as schizophrenia. Cognitive decline is one of the most serious impairments accompanying both AD and schizophrenia. In the present study, the activities of NO donors, slow (spermine NONOate) or fast (DETANONOate) releasers, and selective inhibitor of neuronal nitric oxide synthase N(ω)-propyl-l-arginine (NPLA) were investigated in pharmacological models of schizophrenia and AD. Cognitive impairments were induced by administration of MK-801 or scopolamine and were measured in novel object recognition (NOR) and Y-maze tests. The compounds were investigated at doses of 0.05-0.5 mg/kg. The dose-dependent effectiveness of all the compounds was observed in the NOR test, while only the highest doses of spermine NONOate and NPLA were active in the Y-maze test. DETANONOate was not active in the Y-maze test. The impact of the investigated compounds on motor coordination was tested at doses of 0.5 and 1 mg/kg. Only NPLA at a dose of 1 mg/kg slightly disturbed motor coordination in animals.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Disfunción Cognitiva/tratamiento farmacológico , Donantes de Óxido Nítrico/uso terapéutico , Óxido Nítrico/metabolismo , Nootrópicos/uso terapéutico , Esquizofrenia/tratamiento farmacológico , Enfermedad de Alzheimer/inducido químicamente , Animales , Arginina/análogos & derivados , Arginina/uso terapéutico , Disfunción Cognitiva/inducido químicamente , Maleato de Dizocilpina , Inhibidores Enzimáticos/uso terapéutico , Masculino , Ratones , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Compuestos Nitrosos/uso terapéutico , Prueba de Campo Abierto/efectos de los fármacos , Prueba de Desempeño de Rotación con Aceleración Constante , Esquizofrenia/inducido químicamente , Escopolamina , Espermina/análogos & derivados , Espermina/uso terapéutico
5.
Pharmacol Biochem Behav ; 211: 173300, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34798097

RESUMEN

The spontaneous object recognition (SOR) task is one of the most widely used behavioral protocols to assess visual memory in animals. However, only recently was it shown that nonhuman primates also perform well on this task. Here we further characterized this new monkey recognition memory test by assessing the performance of adult marmosets after an acute systemic administration of two putative amnesic agents: the competitive muscarinic acetylcholine receptor antagonist scopolamine (SCP; 0.05 mg/kg) and the noncompetitive N-methyl-d-aspartate glutamate receptor antagonist MK-801 (0.015 mg/kg). We also determined whether the acetylcholinesterase inhibitor donepezil (DNP; 0.50 mg/kg), a clinically-used cognitive enhancer, reverses memory deficits caused by either drug. The subjects had an initial 10 min sample trial where two identical neutral objects could be explored. After a 6 h retention interval, recognition was based on an exploratory preference for a new rather than familiar object during a 10 min test trial. Both SCP and MK-801 impaired the marmosets' performance on the SOR task, as both objects were explored equivalently. Co-administration of 0.50 mg/kg of DNP reversed the SCP- but not the MK-801-induced memory deficit. These results indicate that cholinergic and glutamatergic pathways mediate object recognition memory in the monkey SOR task.


Asunto(s)
Maleato de Dizocilpina/farmacología , Prueba de Campo Abierto/efectos de los fármacos , Reconocimiento en Psicología/efectos de los fármacos , Escopolamina/farmacología , Acetilcolinesterasa/metabolismo , Animales , Callithrix/metabolismo , Donepezilo/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Haplorrinos/metabolismo , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Antagonistas Muscarínicos/farmacología , Nootrópicos/farmacología , Receptores Muscarínicos/metabolismo
6.
Sci Rep ; 11(1): 22852, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34819526

RESUMEN

Depression and anxiety, two of the most common mental health disorders, share common symptoms and treatments. Most pharmacological agents available to treat these disorders target monoamine systems. Currently, finding the most effective treatment for an individual is a process of trial and error. To better understand how disease etiology may predict treatment response, we studied mice exposed developmentally to the selective serotonin reuptake inhibitor (SSRI) fluoxetine (FLX). These mice show the murine equivalent of anxiety- and depression-like symptoms in adulthood and here we report that these mice are also behaviorally resistant to the antidepressant-like effects of adult SSRI administration. We investigated whether tianeptine (TIA), which exerts its therapeutic effects through agonism of the mu-opioid receptor instead of targeting monoaminergic systems, would be more effective in this model. We found that C57BL/6J pups exposed to FLX from postnatal day 2 to 11 (PNFLX, the mouse equivalent in terms of brain development to the human third trimester) showed increased avoidant behaviors as adults that failed to improve, or were even exacerbated, by chronic SSRI treatment. By contrast, avoidant behaviors in these same mice were drastically improved following chronic treatment with TIA. Overall, this demonstrates that TIA may be a promising alternative treatment for patients that fail to respond to typical antidepressants, especially in patients whose serotonergic system has been altered by in utero exposure to SSRIs.


Asunto(s)
Antidepresivos de Segunda Generación/toxicidad , Antidepresivos Tricíclicos/farmacología , Reacción de Prevención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Fluoxetina/toxicidad , Inhibidores Selectivos de la Recaptación de Serotonina/toxicidad , Tiazepinas/farmacología , Animales , Animales Recién Nacidos , Encéfalo/crecimiento & desarrollo , Conducta Alimentaria/efectos de los fármacos , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Prueba de Campo Abierto/efectos de los fármacos
7.
Lipids Health Dis ; 20(1): 164, 2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789244

RESUMEN

BACKGROUND: Quercetin (QUE) is a flavonol reported with anti-inflammatory and antioxidant activities, and previous results from the group of this study have demonstrated its neuroprotective effect against lipopolysaccharide-induced neuropsychiatric injuries. However, little is known about its potential effect on neuropsychiatric injuries induced or accompanied by metabolic dysfunction of glucose and lipids. METHODS: A nonalcoholic fatty liver disease (NAFLD) rat model was induced via a high-fat diet (HFD), and glucolipid parameters and liver function were measured. Behavioral performance was observed via the open field test (OFT) and the Morris water maze (MWM). The plasma levels of triggering receptor expressed on myeloid cells-1 (TREM1) and TREM2 were measured via enzyme-linked immunosorbent assay (ELISA). The protein expression levels of Synapsin-1 (Syn-1), Synaptatogmin-1 (Syt-1), TREM1 and TREM2 in the hippocampus were detected using western blotting. Morphological changes in the liver and hippocampus were detected by HE and Oil red or silver staining. RESULTS: Compared with the control rats, HFD-induced NAFLD model rats presented significant metabolic dysfunction, hepatocyte steatosis, and impaired learning and memory ability, as indicated by the increased plasma concentrations of total cholesterol (TC) and triglyceride (TG), the impaired glucose tolerance, the accumulated fat droplets and balloon-like changes in the liver, and the increased escaping latency but decreased duration in the target quadrant in the Morris water maze. All these changes were reversed in QUE-treated rats. Moreover, apart from improving the morphological injuries in the hippocampus, treatment with QUE could increase the decreased plasma concentration and hippocampal protein expression of TREM1 in NAFLD rats and increase the decreased expression of Syn-1 and Syt-1 in the hippocampus. CONCLUSIONS: These results suggested the therapeutic potential of QUE against NAFLD-associated impairment of learning and memory, and the mechanism might involve regulating the metabolic dysfunction of glucose and lipids and balancing the protein expression of synaptic plasticity markers and TREM1/2 in the hippocampus.


Asunto(s)
Trastornos de la Memoria/tratamiento farmacológico , Enfermedades Metabólicas/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Quercetina/uso terapéutico , Animales , Western Blotting , Modelos Animales de Enfermedad , Prueba de Tolerancia a la Glucosa , Hígado/patología , Masculino , Glicoproteínas de Membrana/sangre , Trastornos de la Memoria/etiología , Enfermedades Metabólicas/etiología , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Prueba de Campo Abierto/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Inmunológicos/sangre , Receptor Activador Expresado en Células Mieloides 1/sangre
8.
Neurotoxicology ; 87: 156-166, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34597708

RESUMEN

The growing concern surrounding bisphenol A (BPA) has led to increased industrial production and application of its analog bisphenol S (BPS). The goals of this study were: (1) To examine the generational effects in the nematode C. elegans for up to three generations following developmental exposure to BPS (0.1, 1.0, 5.0 and 10.0 µM), and (2) To examine the neurotoxicity and metabolic toxicity in NODEF mouse offspring exposed to BPS (3 µg/kg BW) in utero throughout gestation once/day via oral pipette. First, worms were exposed to BPS developmentally for a single period of 48 hours and then propagated for 2 additional generations. Exposure to 0.1 and 1.0 µM BPS decreased lifespan and the number of progeny with an ability to recover in subsequent generations. In contrast, worms exposed to 5.0 or 10.0 µM BPS exhibited a continuous effect in the second generation, e.g., decreased lifespan and reduced number of progeny. Only worms exposed to 10.0 µM BPS continued to have a significant long-term effect (e.g., decreased lifespan) through the third generation. In addition, worms developmentally exposed to BPS at 5.0 µM and 10.0 µM also showed decreases in body bends. In contrast, worms exposed to 0.1 µM BPS exhibited a significant increase in head thrashes. When the multigenerational effects were examined by exposing worms to BPS for 48 hours developmentally at each generation for three generations, an accumulative effect was observed in worms treated with 0.1 or 1.0 µM BPS for two generations, but not for three generations, suggesting a threshold existed. Worms exposed to either 5.0 or 10.0 µM BPS demonstrated accumulative effects through two and three generations. When the developmental effects of BPS were studied in NODEF mice, offspring exposed gestationally exhibited behavioral deficits at 12, but not at 3, weeks of age. Specifically, female offspring had decreases in working and short-term memories while male offspring showed increases in hyperactivity and anxiety-like behaviors. In summary, this study demonstrates the sex-related effects of BPS in NODEF mouse offspring exposed in utero, along with the generational effects observed in C. elegans.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Sulfonas/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Glucemia/análisis , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Caenorhabditis elegans/crecimiento & desarrollo , Femenino , Fertilidad/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Suspensión Trasera , Longevidad/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones/crecimiento & desarrollo , Prueba de Campo Abierto/efectos de los fármacos , Fenoles/administración & dosificación , Embarazo , Efectos Tardíos de la Exposición Prenatal/psicología , Sulfonas/administración & dosificación
9.
Biomed Pharmacother ; 144: 112303, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34673424

RESUMEN

BACKGROUND: Depressive disorders induced by acute myocardial infarction (AMI) play a pivotal role in the deterioration of cardiac function, and Shuangxinfang (Psycho-cardiology Formula, PCF) was reported to alleviate heart function damage and improve depression-like behavior, but the complex mechanism in such process has not been clarified. METHODS: AMI models were established and PCF was administered in rats. Subjects were then assessed in open field test (OFT) and forced swimming test (FST) recapitulating symptoms of depressive disorder. Afterward, pharmacoproteomic profiling of the hippocampus and peri-infarct border zone (BZ) was performed using a label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) technique, to identify contributing proteins and pathways responsible for myocardial ischemia and behavioral allostasis. Bioinformatics analysis was processed for further investigation, while western blotting was employed for testing dominating proteins to validate proteomic results. RESULTS: Rats in the AMI group showed depression-like behavior in OFT and FST, which was improved by PCF. There were 131 differentially expressed proteins (DEPs) in BZ and 64 proteins in the hippocampus being detected and quantified shared by the sham group, the AMI group, and the PCF group. Subsequently, pertinent pathways and molecular functions were further identified. Altered molecules were discovered to be enriched in the apoptotic process, innate immune response, and NF-κB transcription factor activity in BZ, as well as chemical synaptic transmission, axon, collagen binding, cell adhesion, response to carbohydrate, laminin binding, and cellular response to nitric oxide in the hippocampus. Groups of signal transducers were also able to select multiple pathways, including innate immunity and arginine biosynthesis in the heart, also integrin signaling in the brain. DEPs were intersected from the myocardium and hippocampus to screen out the protein S100A9, which was up-regulated in the AMI group compared with the sham, and showed a down-regulation trend after treatment with PCF. CONCLUSION: Taken together, we present a comprehensive proteomics analysis of rat models with depression post-AMI. Reviewing the literatures concerned, it's hypothesized that macrophage/microglia inflammation mediated by S100A9 might be the pivotal pathogenic process of psycho-cardiology disease, as well as potential mechanisms for the treatment of PCF.


Asunto(s)
Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Calgranulina B/metabolismo , Fármacos Cardiovasculares/farmacología , Depresión/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Hipocampo/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Miocardio/metabolismo , Proteómica , Animales , Cromatografía Líquida de Alta Presión , Cromatografía de Fase Inversa , Depresión/metabolismo , Depresión/psicología , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Hipocampo/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Actividad Motora/efectos de los fármacos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Prueba de Campo Abierto/efectos de los fármacos , Mapas de Interacción de Proteínas , Proteoma , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Función Ventricular Izquierda/efectos de los fármacos
10.
Anesth Analg ; 133(5): 1140-1151, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34673725

RESUMEN

BACKGROUND: Parkinson disease is a chronic and progressive movement disorder characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). The causes of Parkinson disease are not clear but may involve genetic susceptibilities and environmental factors. As in other neurodegenerative disorders, individuals predisposed to Parkinson disease may have an accelerated onset of symptoms following perioperative stress such as anesthesia, surgery, pain, and inflammation. We hypothesized that anesthesia alone accelerates the onset of Parkinson disease-like pathology and symptoms. METHODS: A presymptomatic Parkinson rat model (the protein, DJ-1, encoded by the Park7 gene [DJ-1], PARK7 knockout) was exposed to a surgical plane of isoflurane or 20% oxygen balanced with nitrogen for 2 hours on 3 occasions between 6 and 7 months of age. Acute and long-term motor and neuropathological effects were examined from 7 to 12 months of age in male DJ-1 rats, using the ladder rung, rotarod, and novel object recognition assays, as well as the immunohistochemical localization of tyrosine hydroxylase in dopaminergic neurons in the substantia nigra and ionized calcium-binding adaptor protein-1 (Iba-1) microglial activation in the substantia nigra and hippocampus. RESULTS: In the acute group, after the third anesthetic exposure at 7 months of age, the isoflurane group had a significant reduction in the density of dopaminergic neurons in the SNpc compared to controls. However, this reduction was not associated with increased microglial activation in the hippocampus or substantia nigra. With the ladder rung motor skills test, there was no effect of anesthetic exposure on the total number of foot faults or the ladder rung pattern in the acute group. The rotarod test also detected no differences before and after the third exposure in controls. For the long-term group, immunohistochemical analyses detected no differences in the density of dopaminergic neurons or microglial cells compared to unexposed DJ-1 rats from 8 to 12 months of age. The ladder rung test in the long-term group showed no differences in the total number of foot faults with time and exposure or between ladder rung patterns. The rotarod test detected no significant effect of exposure with time or between groups at any time point. The novel object recognition task in the long-term group revealed no differences in short- or long-term memory or in the number of rearings as a function of exposure. CONCLUSIONS: Multiple isoflurane exposures in this rat model of Parkinson disease transiently enhanced dopaminergic neurodegeneration in the SNpc that resolved over time and had no effects on progression in this Parkinson disease-like phenotype.


Asunto(s)
Anestésicos por Inhalación/toxicidad , Neuronas Dopaminérgicas/efectos de los fármacos , Isoflurano/toxicidad , Degeneración Nerviosa , Trastornos Parkinsonianos/inducido químicamente , Porción Compacta de la Sustancia Negra/efectos de los fármacos , Proteína Desglicasa DJ-1/genética , Animales , Conducta Animal/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Técnicas de Inactivación de Genes , Masculino , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Actividad Motora/efectos de los fármacos , Prueba de Campo Abierto/efectos de los fármacos , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Porción Compacta de la Sustancia Negra/metabolismo , Porción Compacta de la Sustancia Negra/patología , Proteína Desglicasa DJ-1/deficiencia , Ratas Long-Evans , Ratas Transgénicas , Prueba de Desempeño de Rotación con Aceleración Constante , Factores de Tiempo , Tirosina 3-Monooxigenasa/metabolismo
11.
Biomed Pharmacother ; 144: 112280, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34628167

RESUMEN

Metformin is currently used as a first-line drug to treat patients with type 2 diabetes. Previous studies have demonstrated that metformin has antioxidant properties and reduces neuroinflammation and hippocampal neuronal cell loss, which eventually improves memory. Methotrexate (MTX) is an antimetabolite chemotherapeutic agent reported to activate cognitive impairment found in many patients. Moreover, MTX negatively affects the spatial working memory, related to neurogenesis reduction in animal models. Therefore, the present study aimed to investigate the antioxidant effect of metformin on the reduction of memory and neurogenesis caused by MTX. Male Sprague-Dawley rats were divided into four groups: control, MTX, metformin, and MTX+metformin. MTX (75 mg/kg, i.v.) was administered on days 7 and 14. Rats were administered metformin (200 mg/kg, i.p.) for 14 days. Memory was determined using novel object location (NOL) and novel object recognition (NOR) tests. Furthermore, cell cycle arrest was quantified by p21 immunostaining. Levels of neuronal protein expression, scavenging enzymes activity, and malondialdehyde (MDA) level changes in the hippocampus and prefrontal cortex were investigated. Rats receiving only MTX showed memory impairment. Decreases in scavenging enzyme activity and BDNF, DCX, and Nrf2 protein expressions levels were detected in the MTX-treated rats. In addition, MTX significantly increased p21-positive cell numbers and MDA levels. However, these adverse MTX effects were counteracted by co-administration with metformin. These results demonstrate that metformin can improve memory impairments, increase BDNF, DCX and Nrf2 protein expressions and antioxidant capacities, and decrease MDA levels in MTX-treated rats.


Asunto(s)
Conducta Animal/efectos de los fármacos , Hipocampo/efectos de los fármacos , Trastornos de la Memoria/prevención & control , Memoria/efectos de los fármacos , Metformina/farmacología , Neurogénesis/efectos de los fármacos , Nootrópicos/farmacología , Estrés Oxidativo/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Proteína Doblecortina/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Metotrexato , Factor 2 Relacionado con NF-E2/metabolismo , Prueba de Campo Abierto/efectos de los fármacos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Ratas Sprague-Dawley
12.
Biomed Pharmacother ; 144: 112266, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34634555

RESUMEN

BACKGROUND: In the present study, we aimed to investigate the effects of probucol on aging-related hippocampus-dependent cognitive impairment and explore the potential mechanisms. METHODS: D-galactose (100 mg/kg, once daily for 6 weeks) was subcutaneously injected to induce aging in mice. Then the mice were administered with probucol or vehicle once a day for 2 weeks. The hippocampus-related cognition was evaluated with Morris water maze test, novel object recognition test, and contextual fear conditioning test. Moreover, synaptic plasticity was assessed, and RNA-sequencing was applied to further explore the molecular mechanisms. RESULTS: Aging mice induced by D-galactose showed conspicuous learning and memory impairment, which was significantly ameliorated by probucol. Meanwhile, probucol enhanced the spine density and dendritic branches, improved long-term potentiation, and increased the expression of PSD95 of aging mice. Probucol regulated 70 differentially expressed genes compared to D-galactose group, of which 38 genes were upregulated and 32 genes were downregulated. At last, RNA-sequencing results were verified by quantitative reverse transcription-polymerase chain reaction. CONCLUSIONS: Probucol improved learning and memory in aging mice through enhancing synaptic plasticity and regulating gene expression, indicating the potential application of probucol to prevent and treat aging-related disorders.


Asunto(s)
Conducta Animal/efectos de los fármacos , Cognición/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Nootrópicos/farmacología , Probucol/farmacología , Factores de Edad , Animales , Senescencia Celular/efectos de los fármacos , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/psicología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Miedo/efectos de los fármacos , Regulación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Prueba de Campo Abierto/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
13.
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
14.
Neurotoxicology ; 87: 30-42, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34478769

RESUMEN

The mitochondrial inhibitor 3-nitropropionic acid (3-NP) induces excitotoxicity. The authors hypothesized that CTK 01512-2, a recombinant peptide calcium channel N-type blocker, and the TRPA1 antagonist, could show neuroprotective effects. The male Wistar rats received 3-NP [25 mg/kg (i.p.) for 7 days], and a treatment of CTK 01512-2 was delivered intrathecally (i.t.), thrice a week. The neuroprotective effects were evaluated by [18F]FDG MicroPET analysis. The CTK 01512-2 toxin was able to reestablish similar glucose uptakes on the control animals. To detect the neurobehavioral effects from 3-NP, three protocols (6.25, 12.5, 18.75 mg/kg of 3-NP (i.p.), for 3, 4, and 6 days, respectively) were evaluated by performance tests (open field test, walk footprint, elevated plus-maze, Y-maze, and the object recognition test). Important disabilities in the gait of the rats were seen, as well as memory deficits, and anxious behavior in the animals that were treated with all 3-NP protocols. The dose of 18.75 mg/kg (for 3 days) showed the most pronounced behavioral effects and lethality, while the rats treated with 12.5 mg/kg (for 4 days) showed behavioral effects similar to the 6.25 mg/kg dose (for 6 days). The third protocol was then repeated and the rats were treated with the CTK 01512-2 toxin to be evaluated behaviorally again. The recombinant peptide prevented all of the gait-evaluated parameters that were induced by 3-NP at a 6.25 mg/kg dose, which displayed an improvement in the exploratory activities. Overall, these results have reinforced the positive effects of CTK 01512-2 against the behavioral changes that were induced by the mitochondrial inhibitor 3-NP.


Asunto(s)
Bloqueadores de los Canales de Calcio , Fármacos Neuroprotectores , Neurotoxinas , Nitrocompuestos , Propionatos , Animales , Masculino , Ratas , Bloqueadores de los Canales de Calcio/farmacología , Relación Dosis-Respuesta a Droga , Inyecciones Espinales , Fármacos Neuroprotectores/farmacología , Neurotoxinas/antagonistas & inhibidores , Neurotoxinas/toxicidad , Nitrocompuestos/antagonistas & inhibidores , Nitrocompuestos/toxicidad , Prueba de Campo Abierto/efectos de los fármacos , Propionatos/antagonistas & inhibidores , Propionatos/toxicidad , Ratas Wistar , Proteínas Recombinantes , Canal Catiónico TRPA1/antagonistas & inhibidores
15.
Neurotoxicology ; 87: 94-105, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34543681

RESUMEN

Manganese (Mn) is an essential micronutrient but excessive levels induce neurotoxic effects. Increasing evidence suggests a deficit of bioavailable Mn in Huntington disease (HD), an inherited neurodegenerative disease characterized by motor and cognitive disturbances. Previous studies have shown rescue of some molecular HD phenotypes by acute Mn exposure. This study simultaneously examined the potential for chronic Mn exposure to attenuate HD behavioral phenotypes, and for the HD genotype to offer protection against detrimental effects of chronic Mn exposure. In two independent studies a chronic Mn exposure paradigm was implemented in the YAC128 mouse model of HD and behavior was assessed at several timepoints. Study 1 exposed WT and YAC128 mice to twice weekly subcutaneous injections of 0, 5, 15, or 50 mg/kg MnCl[2] tetrahydrate from 12 to 32 weeks of age. A promising protective effect against motor coordination decline in 5 mg/kg MnCl[2] tetrahydrate-treated YAC128 mice was detected. Study 2 thus exposed WT and YAC128 mice to either 0 or 5 mg/kg MnCl[2] tetrahydrate from 12 to 52 weeks of age (with a partial randomized treatment crossover at 31 weeks). The same protective effect was not observed under these conditions at higher statistical power. We report subtle toxicological changes in exploratory behavior and total activity induced by chronic Mn exposure in WT mice only, despite similar total increases in brain Mn in WT and YAC128 mice. Further, chronic Mn treatment resulted in a 10-12 % decrease in striatal NeuN positive cell density in WT mice but not YAC128 mice, despite vehicle cell counts already being reduced compared to WT mice as expected for the HD genotype. The subtle changes observed in specific outcome measures, but not others, following long-term low-level Mn exposure in WT mice delineate the neurobehavioral and neuropathological effects at the threshold of chronic Mn toxicity. We conclude that these chronic low-dose Mn exposures do not significantly rescue behavioral HD phenotypes, but YAC2128 mice are protected against the subtle Mn-induced behavioral changes and decreased striatal neuron density observed in Mn-exposed WT mice.


Asunto(s)
Enfermedad de Huntington/patología , Manganeso/toxicidad , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Fuerza de la Mano , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Masculino , Manganeso/administración & dosificación , Manganeso/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Fármacos Neuroprotectores/administración & dosificación , Prueba de Campo Abierto/efectos de los fármacos , Prueba de Desempeño de Rotación con Aceleración Constante
16.
Neurotoxicology ; 87: 70-85, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34481871

RESUMEN

The number of people with dementia worldwide is estimated at 50 million by 2018 and continues to rise mainly due to increasing aging and population growth. Clinical impact of current interventions remains modest and all efforts aimed at the identification of new therapeutic approaches are therefore critical. Previously, we showed that JM-20, a dihydropyridine-benzodiazepine hybrid molecule, protected memory processes against scopolamine-induced cholinergic dysfunction. In order to gain further insight into the therapeutic potential of JM-20 on cognitive decline and Alzheimer's disease (AD) pathology, here we evaluated its neuroprotective effects after chronic aluminum chloride (AlCl3) administration to rats and assessed possible alterations in several types of episodic memory and associated pathological mechanisms. Oral administration of aluminum to rodents recapitulates several neuropathological alterations and cognitive impairment, being considered a convenient tool for testing the efficacy of new therapies for dementia. We used behavioral tasks to test spatial, emotional- associative and novel object recognition memory, as well as molecular, enzymatic and histological assays to evaluate selected biochemical parameters. Our study revealed that JM-20 prevented memory decline alongside the inhibition of AlCl3 -induced oxidative stress, increased AChE activity, TNF-α and pro-apoptotic proteins (like Bax, caspase-3, and 8) levels. JM-20 also protected against neuronal damage in the hippocampus and prefrontal cortex. Our findings expanded our understanding of the ability of JM-20 to preserve memory in rats under neurotoxic conditions and confirm its potential capacity to counteract cognitive impairment and etiological factors of AD by breaking the progression of key steps associated with neurodegeneration.


Asunto(s)
Cloruro de Aluminio/toxicidad , Benzodiazepinas/farmacología , Trastornos de la Memoria/inducido químicamente , Memoria/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Niacina/análogos & derivados , Cloruro de Aluminio/antagonistas & inhibidores , Animales , Hipocampo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Niacina/farmacología , Prueba de Campo Abierto/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Wistar , Prueba de Desempeño de Rotación con Aceleración Constante
17.
Neurotox Res ; 39(6): 2042-2055, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34499332

RESUMEN

Aluminum is a widespread environmental neurotoxicant that can induce Alzheimer's disease (AD)-like damage, such as neuronal injury and impairment of learning and memory. Several studies have shown that aluminum could reduce the synaptic plasticity, but its molecular mechanism remains unclear. In this study, rats were treated with aluminum maltol (Al(mal)3) to establish a toxic animal model and PMA was used to interfere with the expression of PKC. The Morris water maze and open field test were used to investigate the behavioral changes of the rats. Western blotting and RT-PCR were used to detect the expression levels of NMDAR subunits, PKC and CaMKII. The results showed that Al(mal)3 damaged learning and memory function and reduced anxiety in rats. During this process, the expression of PKC was downregulated and it inhibited the expression of NMDARs through the phosphorylation of CaMKII.


Asunto(s)
Aluminio/toxicidad , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Proteína Quinasa C/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Western Blotting , Relación Dosis-Respuesta a Droga , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Prueba de Campo Abierto/efectos de los fármacos , Proteína Quinasa C/fisiología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de N-Metil-D-Aspartato/fisiología
18.
Biomed Pharmacother ; 143: 112087, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34474339

RESUMEN

Stress during pregnancy is not only detrimental to a woman's own physical and mental health, but can also cause changes in the intrauterine environment and even have an impact on later growth and development, this study was designed to understand the changes of gut microbiota in the maternal and offspring caused by prenatal chronic stress, and to explore the regulatory effect of LBP on gut microbiota, and then to improve the emotional damage caused by prenatal chronic stress in the offspring. A rat model of prenatal chronic stress was made and used LBP to intervene by gavage. Fresh feces of offspring were collected, the concentration of microbial metabolites were tested by ELISA. Illumina MiSeqPE300 sequencing technology was used to determine the sequence of 16S rRNA V3-V4 of microorganisms. On the PND 42, the emotional function of offspring were tested by open-field test (OFT), sucrose preference test (SPT) and tail of suspend test (TST). Results indicated that stress factors increased the plasma corticosterone level of rats during pregnancy and they appeared depressive behaviors. The body weight of offspring during prenatal chronic stress was lower than the control group, and the plasma corticosterone level was increased. Prenatal chronic stress had a significant impact on emotional performance of the offspring on OFT, SPT and TST. Alpha diversity of gut microbiota and microbiota composition in offspring of prenatal chronic stress was attenuated and some relationships existed between these parameters. LBP treatment reduced offspring's plasma corticosterone level and improved their body weight, changed the emotional function, increased the diversity of gut microbiota. Collectively, these findings disclose that prenatal chronic stress not only causes emotional injury on the offspring, but also changes the gut microbiota of the mother and offspring; LBP may regulate the intestinal flora of the mother, then reducing the influence of stress factors on the emotional injury of offspring.


Asunto(s)
Bacterias/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Emociones/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Estrés Psicológico/tratamiento farmacológico , Síntomas Afectivos/etiología , Síntomas Afectivos/microbiología , Síntomas Afectivos/prevención & control , Síntomas Afectivos/psicología , Animales , Bacterias/crecimiento & desarrollo , Eje Cerebro-Intestino/efectos de los fármacos , Enfermedad Crónica , Modelos Animales de Enfermedad , Disbiosis , Femenino , Preferencias Alimentarias/efectos de los fármacos , Masculino , Prueba de Campo Abierto/efectos de los fármacos , Embarazo , Ratas Sprague-Dawley , Estrés Psicológico/complicaciones , Estrés Psicológico/microbiología , Estrés Psicológico/psicología
19.
Biosci Rep ; 41(8)2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34355745

RESUMEN

Traditional Chinese medicine detoxification prescription Chaihu-jia-Longgu-Muli decoction (CLMD) relieves depressive symptoms in patients withdrawing from methamphetamine. In the present study, we assessed the effects of CLMD on methamphetamine withdrawal in rats. A methamphetamine-intoxicated rat model was established. Rats were randomly divided into the control, model, high-dosage, medium-dosage, and low-dosage groups, receiving high, medium, and low doses of CLMD, respectively. Weekly body weight measurements revealed that rats treated with methamphetamine had the lowest body weight. The conditioned place preference (CPP) experiment revealed that methamphetamine-intoxicated rats stayed significantly longer in the drug-paired chamber than the control rats. However, after administering high-dosage CLMD, the amount of time the rats spent in the drug-paired chamber was significantly less than that of the model rats. Our open-field test revealed that the model group had lower crossing and rearing scores than the control group. Additionally, rats that received CLMD treatment exhibited higher crossing and rearing scores than the model rats. Striatal dopamine (DA), 5-hydroxytryptamine (5-HT), and endorphins (ß-EP) and serum interleukin (IL)-1α and IL-2 concentrations were estimated. Rats in the model group had lower striatal DA, 5-HT, and ß-EP and higher serum IL-1α and IL-2 concentrations than those in the control group. High-dosage CLMD administration significantly changed the concentrations of these molecules, such that they approached normal concentrations. In general, CLMD could prevent the development of methamphetamine-induced withdrawal symptoms in rats by increasing the DA, 5-HT, and ß-EP and lowering the IL-1α and IL-2 concentrations.


Asunto(s)
Conducta Animal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central , Condicionamiento Psicológico/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Metanfetamina , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Animales , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Interleucina-1alfa/sangre , Interleucina-2/sangre , Masculino , Prueba de Campo Abierto/efectos de los fármacos , Ratas Sprague-Dawley , Serotonina/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/psicología , betaendorfina/metabolismo
20.
Biol Pharm Bull ; 44(10): 1445-1457, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34349049

RESUMEN

Oxytocin (OXT) -"the love hormone"- has been involved in the anti-depressant activity of some selective serotonin reuptake inhibitors (SSRIs). The exact mechanism underlying the OXT pathway in depression is not fully clear. This study aimed to investigate the effect of OXT analogue, carbetocin (CBT) and the SSRI, escitalopram (ESCIT) on depressive-like behaviors following maternal separation (MS). It is worthy to mention that intranasal CBT has been approved by U.S. Food and Drug Administration (FDA) for Prader-Willi syndrome. Adolescent Wistar albino maternally-separated rats were given CBT, (100 µg/animal/d via inhalation route), and, ESCIT, (20 mg kg-1, per os ( p.o.)) either alone or in combination for 7 d. Repeated 3-h MS demonstrated increased immobility time in forced swim test (FST) and decreased locomotor activity in open field test. MS elevated plasma level of adrenocortico-trophic hormone (ACTH) but notably reduced plasma OXT, with no effect on hippocampal OXT-R expression. Following MS, hippocampal contents of 5-hydroxytryptamine receptors (5HT1A-R), serotonin transporter (SERT) were increased. CBT and ESCIT corrected the behavioral dysfunction in FST and suppressed the high levels of ACTH. Additionally, both treatments boosted OXT level, reduced 5HT1A-R and normalized SERT contents, which reflects increased availability of serotonin. Finally, CBT markedly ameliorated the histopathological damage induced by MS and suppressed the increased glial fibrillary acidic protein. CBT and ESCIT manage depressive-like behavior by positively affecting serotonergic and oxytocinergic systems. Targeting OXT system -using CBT- ameliorated depressive like behaviors induced by maternal separation most probably via enhancing OXT plasma levels, attenuating hormonal ACTH and restoring the expression of hippocampal oxytocin and serotonin mechanisms.


Asunto(s)
Antidepresivos/uso terapéutico , Citalopram/uso terapéutico , Depresión/tratamiento farmacológico , Privación Materna , Oxitocina/análogos & derivados , Hormona Adrenocorticotrópica/sangre , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Masculino , Prueba de Campo Abierto/efectos de los fármacos , Oxitocina/sangre , Oxitocina/uso terapéutico , Ratas , Ratas Wistar , Receptor de Serotonina 5-HT1A/metabolismo , Receptores de Oxitocina/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...