Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
Biochimie ; 191: 51-61, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34454977

RESUMEN

Methylphenidate (MPD), a psychostimulant, is a prescription medicine for treating attention deficit hyperactivity disorder (ADHD). Previously we have shown that moderate doses of MPD enhanced learning and memory while higher doses impaired it. To understand neurochemical mechanisms and receptors involved in memory enhancing and impairing effects of MPD, the present study concerns the effects of these doses of MPD on serotonin, 5-HT1A, GABA, and NMDA receptor mRNA expression in the prefrontal cortex (PFC). We found that low doses (2.5 mg/kg) of MPD improved performance in the water-maze test but higher doses (5 mg/kg) impaired memory retention. Animals showing improved performance had high 5-HT metabolism in the PFC while these levels were not affected in the group treated with higher MPD doses and exhibiting impaired memory. There was downregulation of 5-HT1A receptors in the PFC of rats treated with higher dose MPD, which didn't occur in low dose of MPD treated animals. Further, a decrease in GABAAreceptor mRNA expression occurred in low doses of MPD treated animals and GluN2A expression was reduced in higher doses of MPD treated animals. The findings suggest that memory enhancing doses of MPD increase 5-HT and reduce GABAA receptor mRNA expression in the PFC to release excitatory glutamate neurons from the inhibitory influence of GABA. Conversely, higher dose of MPD downregulates 5-HT1A receptor mRNA expression to enhance inhibitory GABA influence on glutamate neurons and impair cognitive performance. The findings show an important role of 5-HT1A heteroreceptors in the PFC for improving therapeutic use of MPD and developing novel cognitive enhancers.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Memoria/efectos de los fármacos , Metilfenidato/farmacología , Corteza Prefrontal/metabolismo , Receptor de Serotonina 5-HT1A/biosíntesis , Receptores de GABA-A/biosíntesis , Receptores de N-Metil-D-Aspartato/biosíntesis , Serotonina/metabolismo , Animales , Masculino , Ratas , Ratas Wistar
2.
Neurotoxicology ; 85: 245-253, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34111468

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects many older people around the world. Numerous studies are underway to evaluate the protective effects of natural products in AD. Alpha-linoleic acid (ALA) is an essential unsaturated fatty acid that exhibits neuroprotective outcomes in rat models of ischemic stroke and Parkinson's disease. This research aimed to investigate the effect of ALA on oxidative stress, neuroinflammation, neuronal death, and memory deficit induced by amyloid-beta (Aß) peptide. After intrahippocampal injection of Aß1-42, rats received ALA (150 µg/kg, subcutaneously) for 14 consecutive days. ALA decreased the levels of malondialdehyde and nitric oxide, enhanced glutathione content, and increased the activity of catalase in the hippocampus of the rat model of AD. It also reduced the expression of tumor necrosis factor-α, interleukin-1ß, interleukin-6, nuclear factor-kappa B, and N-methyl-d-aspartate receptor subunits NR2A and NR2B mRNAs in the hippocampus, prevented the neuronal loss in the CA1 region, and enhanced the expression of α7 nicotinic acetylcholine receptor. In addition, ALA allowed Aß1-42-injected rats to spend less time and distance to reach the hidden platform in the Morris water maze test and to swim longer in the target quadrant. We concluded that ALA reduces the biochemical, molecular, histological, and behavioral changes caused by Aß1-42 and it may be an effective option for treating AD.


Asunto(s)
Hipocampo/efectos de los fármacos , Trastornos de la Memoria/prevención & control , Estrés Oxidativo/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Ácido alfa-Linolénico/uso terapéutico , Receptor Nicotínico de Acetilcolina alfa 7 , Péptidos beta-Amiloides/toxicidad , Animales , Expresión Génica , Hipocampo/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/fisiología , Fragmentos de Péptidos/toxicidad , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/biosíntesis , Receptores de N-Metil-D-Aspartato/genética , Ácido alfa-Linolénico/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/biosíntesis , Receptor Nicotínico de Acetilcolina alfa 7/genética
3.
Neurotox Res ; 39(3): 634-644, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33464538

RESUMEN

Aluminum demonstrates clear neurotoxicity and can cause Alzheimer's disease (AD)-like symptoms, including cognitive impairment. One toxic effect of aluminum is a decrease in synaptic plasticity, but the specific mechanism remains unclear. In this study, PC12 cells were treated with Al(mal)3 to construct a toxic cell model. (S)-3,5-Dihydroxyphenylglycine (DHPG), α-methyl-4-carboxyphenylglycine (MCPG), and mGluR1-siRNA were used to interfere with the expression of metabotropic glutamate receptor subtype 1 (mGluR1). Polymerase chain reaction and western blotting were used to investigate the expression of mGluR1, protein kinase C (PKC), and N-methyl-D-aspartate receptor (NMDAR) subunits. ELISA was used to detect PKC enzyme activity. In PC12 cells, mRNA and protein expressions of PKC and NMDAR subunits were inhibited by Al(mal)3. Aluminum may further regulate the expression of NMDAR1 and NMDAR2B through mGluR1 to regulate PKC enzyme activity, thereby affecting learning and memory functions. Furthermore, the results implied that the mGluR1-PKC-NMDAR signaling pathway may predominately involve positive regulation. These findings provide new targets for studying the neurotoxic mechanism of aluminum.


Asunto(s)
Aluminio/toxicidad , Proteína Quinasa C/biosíntesis , Receptores de Glutamato Metabotrópico/biosíntesis , Receptores de N-Metil-D-Aspartato/biosíntesis , Animales , Expresión Génica , Glicina/análogos & derivados , Glicina/farmacología , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Células PC12 , Ratas , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
4.
Neurosci Lett ; 721: 134819, 2020 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-32032749

RESUMEN

It is widely reported that drug addiction involves the strengthening of specific reward circuits through N-methyl-d-aspartic acid receptor (NMDAR)-dependent synaptic potentiation, and several lines of evidence strongly implicate NMDA receptor 2 (NR2) subunits in drug abuse. To explore the potential mechanism of heroin dependence, this study examined changes in the expression levels of NR2 subunits NR2A-D in the prelimbic (PL) region of the medial prefrontal cortex (mPFC) after repeated heroin administration and subsequent abstinence. The conditioned place preference (CPP) test confirmed successful induction of heroin dependence and withdrawal. Western blotting and qRT-PCR revealed no differences in NR2A subunit expression among heroin-exposure, heroin-withdrawal, and control group rats; in contrast, expression of NR2B was significantly higher in the heroin-exposure group, whereas expression levels of NR2C and NR2D were significantly higher in the heroin-withdrawal group relative to the controls. Further studies are needed to identify the functional significance based on alterations of NR2 subunits.


Asunto(s)
Dependencia de Heroína/metabolismo , Heroína/efectos adversos , Corteza Prefrontal/metabolismo , Receptores de N-Metil-D-Aspartato/biosíntesis , Síndrome de Abstinencia a Sustancias/metabolismo , Animales , Expresión Génica , Dependencia de Heroína/genética , Masculino , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Síndrome de Abstinencia a Sustancias/genética
5.
Neurol Res ; 42(2): 108-117, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31941414

RESUMEN

Objective: The possible effect of NMDAR (N-methyl-D-aspartate receptor)-NMNAT1/2 (nicotinamide/nicotinic acid mono-nucleotide adenylyltransferase) signaling pathway on the neuronal cell damage and cognitive impairment of aged rats anesthetized by sevoflurane was explored.Methods: Adult male Wistar rats were selected and divided into Control, Sevo (Sevoflurane), Sevo+DCS (NMDAR agonist D-cycloserine) 30 mg/kg, Sevo+DCS 100 mg/kg, and Sevo+DCS 200 mg/kg groups. Morris water maze and fear conditioning text were used to observe cognitive function changes of rats. The inflammatory cytokines were determined by quantitative real-time polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA) assay, neuronal apoptosis by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labelling (TUNEL) staining and MDAR-NMNAT1/2 pathway-related proteins by Western blotting.Results: The longer escape latency, decreased platform crossing times and reduced staying time spent in platform quadrant were found in rats from Sevo group, with decreased percentage of freezing time in contextual test and tone cued test; and meanwhile, these rats had increased inflammatory cytokines (interleukin (IL)-1ß, tumor necrosis factor (TNF-α), IL-6, and IL-8) and neuronal apoptosis, but declined expressions of MDAR-NMNAT1/2 pathway-related proteins. However, the above changes were exhibited an opposite tendency in those Sevo rats treated with different concentrations of DCS (including 30, 100, and 200 mg/kg, respectively). Particularly, the improving effect of low-dose DCS on each aspect in aged rats was better than high-dose ones.Conclusion: Activation of NMDAR-NMNAT1/2 signaling pathway could not only reduce neuronal apoptosis, but also alleviate sevoflurane-induced neuronal inflammation and cognitive impairment in aged rats.


Asunto(s)
Disfunción Cognitiva/prevención & control , Cicloserina/farmacología , Neuronas/patología , Nicotinamida-Nucleótido Adenililtransferasa/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Sevoflurano/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Hipocampo/patología , Masculino , Aprendizaje por Laberinto/fisiología , Nicotinamida-Nucleótido Adenililtransferasa/biosíntesis , Ratas , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/biosíntesis , Transducción de Señal/fisiología
6.
Mol Cell Biochem ; 464(1-2): 73-81, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31732832

RESUMEN

Sepsis-associated acute kidney injury (AKI) is a life threatening condition with high morbidity and mortality. The pathogenesis of AKI is associated with apoptosis. In this study, we investigated the effects of ligustrazine (LGZ) on experimental sepsis-associated AKI in mice. Sepsis-associated AKI was induced in a mice model using cecal ligation and puncture (CLP) method. Mice were administered LGZ (10, 30, and 60 mg/kg) via tail vein injection 0.5 h before CLP surgery. Mice survival was evaluated. Renal water content was detected. Urine samples were collected for ELISA of Kim1. Kidneys were collected for nucleic acid analysis and histological examination. Pathological assessment was used to determine the effect of LGZ on sepsis-associated AKI. Caspase-3 expression in kidney was assessed by immunohistochemistry. Renal NMDAR1 level was also determined. Treatment of LGZ improved mice survival rate; the effect was significant when administered at a high LGZ dose (60 mg/kg). Renal water content of mice undergoing CLP was significantly reduced by LGZ treatment. Both middle-dose and high-dose LGZ treatments reduced urine Kim1 level in sepsis-associated AKI mice. The severity of AKI in septic mice was reduced by middle-dose and high-dose LGZ administration. Immunohistochemical analysis revealed decreased caspase-3 and NMDAR1 levels in the kidney following middle-dose and high-dose LGZ treatments. RT-PCR assay showed a significant reduction in NMDAR1 mRNA expression in the kidney of middle-dose and high-dose LGZ-treated mice. LGZ exhibited protective effects against sepsis-associated AKI in mice, possibly via downregulation of renal NMDAR1 expression and its anti-apoptotic action by inhibiting caspase-3.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Caspasa 3/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Pirazinas/farmacología , Receptores de N-Metil-D-Aspartato/biosíntesis , Sepsis/tratamiento farmacológico , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Sepsis/metabolismo , Sepsis/patología
7.
Psychopharmacology (Berl) ; 236(9): 2687-2697, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31053935

RESUMEN

RATIONALE: Antipsychotics exert therapeutic effects by modulating various cellular signalling pathways and several types of receptors, including PKA- and GSK3ß-mediated signalling pathways, and NMDA receptors. The ventral midbrain, mainly containing the ventral tegmental area (VTA) and substantia nigra (SN), are the nuclei with dopamine origins in the brain, which are also involved in the actions of antipsychotics. Whether antipsychotics can modulate these cellular pathways in the ventral midbrain is unknown. OBJECTIVE: This study aims to investigate the effects of antipsychotics, including aripiprazole (a dopamine D2 receptor (D2R) partial agonist), bifeprunox (a D2R partial agonist), and haloperidol (a D2R antagonist) on the PKA- and GSK3ß-mediated pathways and NMDA receptors in the ventral midbrain. METHODS: Male rats were orally administered aripiprazole (0.75 mg/kg, t.i.d. (ter in die)), bifeprunox (0.8 mg/kg, t.i.d.), haloperidol (0.1 mg/kg, t.i.d.) or vehicle for either 1 week or 10 weeks. The levels of PKA, p-PKA, Akt, p-Akt, GSK3ß, p-GSK3ß, Dvl-3, ß-catenin, and NMDA receptor subunits in the ventral midbrain were assessed by Western Blots. RESULTS: The results showed that chronic antipsychotic treatment with aripiprazole selectively increased PKA activity in the VTA. Additionally, all three drugs elevated the activity of the Akt-GSK3ß signalling pathway in a time-dependent manner, while only aripiprazole stimulated the Dvl-3-GSK3ß-ß-catenin signalling pathway in the SN. Furthermore, chronic administration with both aripiprazole and haloperidol decreased the expression of NMDA receptors. CONCLUSION: This study suggests that activating PKA- and GSK3ß-mediated pathways and downregulating NMDA receptor expression in the ventral midbrain might contribute to the clinical effects of antipsychotics.


Asunto(s)
Antipsicóticos/administración & dosificación , Proteínas Quinasas Dependientes de AMP Cíclico/biosíntesis , Glucógeno Sintasa Quinasa 3 beta/biosíntesis , Mesencéfalo/efectos de los fármacos , Mesencéfalo/metabolismo , Receptores de N-Metil-D-Aspartato/biosíntesis , Animales , Antipsicóticos/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
8.
Neuromolecular Med ; 21(2): 132-142, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30784006

RESUMEN

Epigenetic agents, such as neonatal isolation during neurodevelopmental period of life, can change various regions of the brain. It may further induce psychological disorders such as autistic-like phenomena. This study indicated the role of chronic increased anterior cingulate cortex (ACC) output on alteration of caudate putamen (CPu) as a main behavior regulator region of the brain in adult maternal deprived (MD) rats. For making an animal model, neonates were isolated from their mothers in postnatal days (PND 1-10, 3 h/day). Subsequently, they bilaterally received pLenti-CaMKIIa-hChR2 (H134R)-mCherry-WPRE virus in ACC area via stereotaxic surgery in PND50. After 22 days, these regions were exposed to blue laser (473 nm) for six consecutive days (15 min/day). Then, behavioral deficits were tested and were compared with control group in the following day. Animals were immediately killed and their brains were prepared for tissue processing. Results showed that neonatal isolation induces autistic-like behaviors and leads to overexpression of NMDAR1 and Nox2-gp91phox proteins and elevation of catalase activity in the CPu regions of the adult offspring compared with control group. Chronic optogenetic stimulation of ACC neurons containing (ChR2+) led to significant reduction in the appearance of stereotypical behavior and alien-phobia in MD rats. The amount of NMDAR1 and Nox2-gp91phox expression and the catalase activity in CPu were reduced after this treatment. Therefore, autistic-like behavior seems to be related with elevation of NMDAR1 and Nox2-gp91phox protein levels that enhance the effect of glutamatergic projection on CPu regions. Optogenetic treatment also could ameliorate behavioral deficits by modulating these protein densities.


Asunto(s)
Trastorno Autístico , Núcleo Caudado , Giro del Cíngulo , Privación Materna , Optogenética , Putamen , Animales , Femenino , Masculino , Ratas , Animales Recién Nacidos , Trastorno Autístico/fisiopatología , Trastorno Autístico/terapia , Catalasa/biosíntesis , Catalasa/genética , Núcleo Caudado/fisiopatología , Modelos Animales de Enfermedad , Genes Reporteros , Vectores Genéticos/administración & dosificación , Giro del Cíngulo/fisiopatología , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Microinyecciones , NADPH Oxidasa 2/biosíntesis , NADPH Oxidasa 2/genética , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Fobia Social , Putamen/fisiopatología , Distribución Aleatoria , Receptores de N-Metil-D-Aspartato/biosíntesis , Receptores de N-Metil-D-Aspartato/genética , Conducta Social , Estrés Psicológico , Proteína Fluorescente Roja
9.
Neuroreport ; 29(18): 1564-1570, 2018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30371539

RESUMEN

The current research probed into the effects of green tea polyphenols (GTPs) on ethanol-induced spatial learning and memory impairments and inquired the potential molecular mechanism in rats. Thirty 8-week-old male Sprague-Dawley rats were randomly divided into three groups. The control group (control, n=10), ethanol group (ethanol, n=10), and GTPs intervention group (GTP, n=10) received gavage administration of saline, ethanol, and ethanol-GTP solution, respectively, for 8 weeks. Morris water maze was applied to assess the spatial learning and memory function of rats in each group at the last week of treatment. There was no dramatic change in body weight of rats in the different groups. Compared with rats in the control group, 8-week ethanol gavaged rats showed increased escape latency period and decreased time in the target quadrant. Moreover, 8-week ethanol gavage decreased the density of pyramidal layer neurons, expression of NMDAR1, and CREB phosphorylation in the hippocampus region. In contrast, GTP intervention decreased escape latency period and increased the time in the target quadrant, the density of pyramidal layer neurons, expression of NMDAR1, and CREB phosphorylation in the hippocampus region. The current findings indicated that GTP intervention can improve ethanol-induced spatial learning and memory impairments in rats after ethanol withdrawal, which is related to the upregulated density of pyramidal layer neurons, expression of NMDAR1, and CREB phosphorylation in the hippocampus region.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Etanol/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Polifenoles/farmacología , Receptores de N-Metil-D-Aspartato/biosíntesis , Animales , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Té/química
10.
J Neurochem ; 147(5): 609-625, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30062698

RESUMEN

Vaccinia-related kinase 2 (VRK2) is a serine/threonine kinase that belongs to the casein kinase 1 family. VRK2 has long been known for its relationship with neurodegenerative disorders such as schizophrenia. However, the role of VRK2 and the substrates associated with it are unknown. Dysbindin is known as one of the strong risk factors for schizophrenia. The expression of dysbindin is indeed significantly reduced in schizophrenia patients. Moreover, dysbindin is involved in neurite outgrowth and regulation of NMDA receptor signaling. Here, we first identified dysbindin as a novel interacting protein of VRK2 through immunoprecipitation. We hypothesized that dysbindin is phosphorylated by VRK2 and further that this phosphorylation plays an important role in the function of dysbindin. We show that VRK2 phosphorylates Ser 297 and Ser 299 of dysbindin using in vitro kinase assay. In addition, we found that VRK2-mediated phosphorylation of dysbindin enhanced ubiquitination of dysbindin and consequently resulted in the decrease in its protein stability through western blotting. Over-expression of VRK2 in human neuroblastoma (SH-SY5Y) cells reduced neurite outgrowth induced by retinoic acid. Furthermore, a phosphomimetic mutant of dysbindin alleviated neurite outgrowth and affected surface expression of N-methyl-d-aspartate 2A, a subunit of NMDA receptor in mouse hippocampal neurons. Together, our work reveals the regulation of dysbindin by VRK2, providing the association of these two proteins, which are commonly implicated in schizophrenia. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Asunto(s)
Disbindina/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Estabilidad Proteica , Animales , Línea Celular , Disbindina/genética , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Mutación/fisiología , Neuritas/efectos de los fármacos , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/farmacología , Receptores de N-Metil-D-Aspartato/biosíntesis , Tretinoina/farmacología , Ubiquitinación
11.
Am J Physiol Lung Cell Mol Physiol ; 315(3): L404-L421, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29722562

RESUMEN

Endogenous glutamate (Glu) release and N-methyl-d-aspartate (NMDA) receptor (NMDAR) activation are associated with lung injury in different animal models. However, the underlying mechanism is unclear. Bone marrow-derived mesenchymal stem cells (BM-MSCs), which show potential use for immunomodulation and tissue protection, play a protective role in pulmonary fibrosis (PF) process. Here, we found the increased Glu release from the BM cells of bleomycin (BLM)-induced PF mice in vivo. BLM stimulation also increased the extracellular Glu in BM-MSCs via the antiporter system xc- in vitro. The gene expression of each subunit of NMDAR was detected in BM-MSCs. NMDAR activation inhibited the proliferation, migration, and paracrine function of BM-MSCs in vitro. BM-MSCs were derived from male C57BL/6 mice, transfected with lentiviral vectors carrying the enhanced green fluorescence protein gene, pretreated with NMDA, and transplanted into the female recipient mice that were intratracheally injected with BLM to induce PF. Transplantation of NMDA-pretreated BM-MSCs significantly aggravated PF as compared with that in the normal BM-MSCs transplantation group. The sex determination gene Y chromosome and green fluorescence protein genes of BM-MSCs were detected to observe BM-MSCs homing in the fibrotic lungs. Moreover, NMDAR activation inhibited BM-MSC migration by downregulating the stromal cell-derived factor-1/C-X-C chemokine receptor type 4 signaling axis. NMDAR activation aggravated the transforming growth factor-ß1-induced extracellular matrix production in alveolar epithelial cells and fibroblasts through the paracrine effects of BM-MSCs. In summary, these findings suggested that NMDAR activation-mediated Glu excitotoxicity induced by BLM in BM-MSCs abolished the therapeutic effects of normal BM-MSCs transplantation on BLM-induced PF.


Asunto(s)
Bleomicina/efectos adversos , Células de la Médula Ósea/metabolismo , Ácido Glutámico/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Fibrosis Pulmonar , Receptores de N-Metil-D-Aspartato/biosíntesis , Animales , Bleomicina/farmacología , Células de la Médula Ósea/patología , Movimiento Celular , Proliferación Celular , Regulación de la Expresión Génica , Masculino , Células Madre Mesenquimatosas/patología , Ratones , Comunicación Paracrina , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/prevención & control
12.
Neurochem Int ; 118: 73-81, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29704590

RESUMEN

Evidences have indicated a high degree of comorbidity of alcoholism and depression. N-acetylcysteine (NAC) has shown its clinical efficiency in the treatment of several psychiatric disorders and is identified as a multi-target acting drug. The ability of NAC to prevent alcohol abstinence-induced depression-like effects and underlying mechanism(s) have not been adequately addressed. This study was aimed to investigate the beneficial effects of NAC in the alcohol abstinence-induced depression developed following long-term voluntary alcohol intake. For evaluation of the effects of NAC, Sprague-Dawley rats were enabled to voluntary drinking of 4.5%, 7.5% and 9% v/v alcohol for fifteen days. NAC (25, 50, and 100 mg/kg) and fluoxetine (5 mg/kg) were injected intraperitoneally for three consecutive days during the alcohol abstinence period on the days 16, 17, 18. The behavioral studies were conducted employing forced swim test (FST), and tail suspension test (TST) on day 18 to determine the effects of N-acetylcysteine and fluoxetine in the ethanol withdrawal induced-depression. Blood alcohol concentration, alcohol biomarkers like SGPT, SGOT, ALP, GGT, and MCV were estimated by using commercially available kits. Serotonin concentrations were measured in the plasma, hippocampus and pre-frontal cortex using the rat ELISA kit. The expression of GRIN1, GRIN2A, GRIN2B genes for the N-methyl d-aspartate receptors (NMDAR) subunits in the hippocampus and the prefrontal cortex were also examined by reverse-transcription quantitative polymerase chain reaction. The results revealed that alcohol abstinence group depicted increased immobility time in FST and TST. Further, NAC exerted significant protective effect at the doses 50 mg/kg and 100 mg/kg, but 25 mg/kg showed insignificant protection against alcohol abstinence-induced depression. The increased level of biochemical parameters following ethanol abstinence were also reversed by NAC at the dose of 100 mg/kg. The significant reversal effect of NAC on the serotonin level following alcohol abstinence was greater in the hippocampus as compared to the third-day alcohol withdrawal group. The increased expression levels of GRIN2A and GRIN2B following ethanol abstinence were reversed with a higher dose of NAC (100 mg/kg) treatment. In conclusion, the results of the study reveal that NAC has remarkable protective effects in the alcohol abstinence-induced depression by modulating alcohol markers, serotonin levels and GRIN2A, GRIN2B gene expression of NMDAR signaling pathway in rats.


Asunto(s)
Acetilcisteína/uso terapéutico , Abstinencia de Alcohol/psicología , Depresión/metabolismo , Depresión/psicología , Receptores de N-Metil-D-Aspartato/biosíntesis , Acetilcisteína/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Depresión/tratamiento farmacológico , Depresión/genética , Relación Dosis-Respuesta a Droga , Expresión Génica , Masculino , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
13.
Pain Res Manag ; 2018: 7436060, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29623145

RESUMEN

We constructed a water-soluble lipopolymer (WSLP) as a nonviral gene carrier to deliver siRNA targeting NR2B. The cytotoxicity and serum stability of WSLP loaded with siRNA were evaluated, and the knockdown efficiency of WSLP/NR2B-siRNA in PC12 cells was examined. The results showed that WSLP could protect the loading siRNAs from enzymatic degradation in serum and exhibit low cytotoxicity to cells. After transfection, WSLP/NR2B-siRNA complexes reduced the NR2B transcriptional level by 50% and protein level by 55% compared to control siRNA. Moreover, 3 days after intrathecal injection of WSLP/NR2B-siRNA complexes into rats, the NR2B protein expression decreased significantly to 58%, compared to control treatment (p < 0.01). Injection of WSLP with scrambled siRNA or of polyethylenimine (PEI) with NR2B-siRNA did not show this inhibitory effect. Additionally, injection of WSLP/NR2B-siRNA complexes significantly relieved inflammatory pain in rats at 3, 4, and 5 days with reduced MWT and decreased TWL scores, while injection of WSLP with scrambled siRNA or of PEI with NR2B-siRNA did not. These results demonstrated that WSLP can efficiently deliver siRNA targeting NR2B to PC12 cells and relieve pain in rats with chronic inflammatory pain.


Asunto(s)
Dolor Crónico/terapia , Terapia Genética/métodos , Polímeros/uso terapéutico , ARN Interferente Pequeño/administración & dosificación , Receptores de N-Metil-D-Aspartato/genética , Animales , Dolor Crónico/etiología , Dolor Crónico/patología , Dolor Crónico/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Adyuvante de Freund/toxicidad , Hiperalgesia/etiología , Hiperalgesia/terapia , Inflamación/inducido químicamente , Inflamación/complicaciones , Masculino , Células PC12 , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Polímeros/administración & dosificación , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/biosíntesis , Receptores de N-Metil-D-Aspartato/metabolismo , Asta Dorsal de la Médula Espinal/efectos de los fármacos , Asta Dorsal de la Médula Espinal/metabolismo , Transfección
14.
Neurochem Res ; 43(5): 1047-1057, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29574667

RESUMEN

Exercise and low-fat diets are common lifestyle modifications used for the treatment of hypertension besides drug therapy. However, unrestrained low-fat diets may result in deficiencies of low-unsaturated fatty acids and carry contingent risks of delaying neurodevelopment. While aerobic exercise shows positive neuroprotective effects, it is still unclear whether exercise could alleviate the impairment of neurodevelopment that may be induced by certain low-fat diets. In this research, developing spontaneously hypertensive rats (SHR) were treated with chronic swimming exercise and/or a low-soybean-oil diet for 6 weeks. We found that performance in the Morris water maze was reduced and long-term potentiation in the hippocampus was suppressed by the diet, while a combination treatment of exercise and diet alleviated the impairment induced by the specific low-fat diet. Moreover, the combination treatment effectively increased the expression of brain-derived neurotrophic factor (BDNF) and N-methyl-D-aspartic acid receptor (NMDAR), which were both down-regulated by the low-soybean-oil diet in the hippocampus of developing SHR. These findings suggest that chronic swimming exercise can ameliorate the low-soybean-oil diet-induced learning and memory impairment in developing SHR through the up-regulation of BDNF and NMDAR expression.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Dieta con Restricción de Grasas/efectos adversos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/psicología , Condicionamiento Físico Animal/fisiología , Memoria Espacial/fisiología , Natación/fisiología , Sinapsis/efectos de los fármacos , Anhedonia/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Masculino , Plasticidad Neuronal , Desempeño Psicomotor/efectos de los fármacos , Desempeño Psicomotor/fisiología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de N-Metil-D-Aspartato/biosíntesis , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Aceite de Soja , Regulación hacia Arriba
15.
Pharmacol Biochem Behav ; 167: 1-8, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29421366

RESUMEN

The use of opioid analgesics to treat non-cancer pain has increased over the years. Many chronic pain patients suffer from numerous adverse effects, such as reduced quality of life, development of dependence, and cognitive impairments. Cognitive processes are regulated by several systems, one of which involves growth hormone (GH) and its secondary mediator insulin-like growth factor-1 (IGF-1), but also glutamatergic transmission, including receptors such as the N-methyl-d-aspartate (NMDA)-receptor complex. In the laboratory, repeated injections are commonly used to establish animal models of long-term or chronic drug exposure. However, in the present study, we aimed to mimic a more human dose regimen using constant drug delivery provided by mini-osmotic pumps implanted subcutaneously in male Sprague Dawley rats. After developing opioid tolerance the cognitive function of rats was studied. Spatial learning and memory capabilities were evaluated using the rat Morris water maze (MWM). Moreover, gene expression related to the GH/IGF-1-axis and the NMDA-receptor system was analyzed using quantitative PCR (qPCR) and plasma levels of IGF-1 were assessed using the ELISA technique. Our results demonstrate that rats exposed to morphine for 27 days display memory impairments in the MWM probe trial. However, the behavioral effects of chronic morphine treatment were not accompanied by any significant differences in terms of mRNA expression or IGF-1 plasma concentration. The animal model used in this study provides a simple and suitable way to investigate the behavioral and neurochemical effects of chronic opioid treatment similar to the exposure seen in human pain patients.


Asunto(s)
Trastornos de la Memoria/inducido químicamente , Morfina/administración & dosificación , Morfina/farmacología , Memoria Espacial/efectos de los fármacos , Animales , Proteínas Portadoras/biosíntesis , Tolerancia a Medicamentos , Bombas de Infusión Implantables , Infusiones Subcutáneas , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Factor II del Crecimiento Similar a la Insulina/biosíntesis , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/metabolismo , Ósmosis , Dimensión del Dolor/efectos de los fármacos , Ratas , Receptores de N-Metil-D-Aspartato/biosíntesis
16.
Mol Biol (Mosk) ; 51(5): 870-880, 2017.
Artículo en Ruso | MEDLINE | ID: mdl-29116075

RESUMEN

Profiles of alternative mRNA isoforms have been determined in three brain regions of rats from an aggressive and a tame line selected for 74 generations. Among 2319 genes with alternatively spliced exons, approximately 84% were confirmed by analyzing public databases. Based on Gene Ontology-guided clustering of alternatively spliced genes, it has been found that the sample was enriched in synapse-specific genes (FDR < 10^(-17)). Patterns of gene expression in the brains of animals with genetically determined high or low aggression were more frequently found to differ in the use of alternatively spliced exons than in animals environmentally conditioned for increased or lowered propensity to aggression. For the Adcyap1r1 gene, five alternatively spliced mRNA isoforms have been represented differentially in aggressive animals. A detailed analysis of the gene that encodes glutamate ionotropic receptor NMDA type subunit 1 (Grin1) has confirmed significant differences in the levels of its alternatively spliced isoforms in certain brain regions of tame and aggressive rats. These differences may affect the behavior in rats genetically selected for aggression levels.


Asunto(s)
Agresión , Empalme Alternativo , Encéfalo/metabolismo , ARN Mensajero/metabolismo , Receptores de N-Metil-D-Aspartato/biosíntesis , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/biosíntesis , Animales , Masculino , ARN Mensajero/genética , Ratas , Receptores de N-Metil-D-Aspartato/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Selección Artificial
17.
J Comp Neurol ; 525(5): 1192-1205, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27650492

RESUMEN

Cervical spinal hemisection at C2 (SH) removes premotor drive to phrenic motoneurons located in segments C3-C5 in rats. Spontaneous recovery of ipsilateral diaphragm muscle activity is associated with increased phrenic motoneuron expression of glutamatergic N-methyl-D-aspartate (NMDA) receptors and decreased expression of α-amino-3-hydroxy-5-methylisoxazole-4-proprionic acid (AMPA) receptors. Glutamatergic receptor expression is regulated by tropomyosin-related kinase receptor subtype B (TrkB) signaling in various neuronal systems, and increased TrkB receptor expression in phrenic motoneurons enhances recovery post-SH. Accordingly, we hypothesize that recovery of ipsilateral diaphragm muscle activity post-SH, whether spontaneous or enhanced by adenoassociated virus (AAV)-mediated upregulation of TrkB receptor expression, is associated with increased expression of glutamatergic NMDA receptors in phrenic motoneurons. Adult male Sprague-Dawley rats underwent diaphragm electromyography electrode implantation and SH surgery. Rats were injected intrapleurally with AAV expressing TrkB or GFP 3 weeks before SH. At 14 days post-SH, the proportion of animals displaying recovery of ipsilateral diaphragm activity increased in AAV-TrkB-treated (9/9) compared with untreated (3/5) or AAV-GFP-treated (4/10; P < 0.027) animals. Phrenic motoneuron NMDA NR1 subunit mRNA expression was approximately fourfold greater in AAV-TrkB- vs. AAV-GFP-treated SH animals (P < 0.004) and in animals displaying recovery vs. those not recovering (P < 0.005). Phrenic motoneuron AMPA glutamate receptor 2 (GluR2) subunit mRNA expression decreased after SH, and, albeit increased in animals displaying recovery vs. those not recovering, levels remained lower than control. We conclude that increased phrenic motoneuron expression of glutamatergic NMDA receptors is associated with spontaneous recovery after SH and enhanced recovery after AAV-TrkB treatment. J. Comp. Neurol. 525:1192-1205, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Diafragma/inervación , Neuronas Motoras/metabolismo , Receptores de N-Metil-D-Aspartato/biosíntesis , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Animales , Modelos Animales de Enfermedad , Captura por Microdisección con Láser , Masculino , Nervio Frénico/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor trkB/metabolismo
18.
Behav Brain Res ; 318: 36-44, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27780721

RESUMEN

Molecular mechanisms of depression-like pathophysiology in female rodent models are less reported compared to males, despite its higher prevalence in human females. Moreover, the stress-response in brain circuitries including reward and cognition circuitries varies with age or hormonal status of the females. So, to understand the stress-induced mood and cognitive disorders in intact females (with ovaries) and ovariectomized (OVX) females, we studied changes in mouse hippocampus, a functionally heterogeneous neural structure involved in both affective and cognitive behaviors. Here, we used a 6-day Chronic Unpredictable Stress (CUS) paradigm in mice to induce depression and related mood disorders. Interestingly, intact females and OVX females showed difference in mood disorder sub-phenotypes to CUS. Similar to an earlier report of CUS affecting the critical reward circuitry structure the nucleus accumbens differently in females with and without ovaries, cognitive behavior in intact females and OVX females also responded differentially to CUS, as evident from Morris Water Maze (MWM) test results. We report that the presence or absence of ovarian hormones, particularly the estrogen, has a significant impact in altering the hippocampus related spatial memory and affective behavior, in females. Our results also illustrate that estrogen administration improves both reward and cognitive behavior, and plays a significant role in alleviating stress-induced despair behavior and enhancing spatial reference memory following a brief 6-day stressful paradigm. Further, it also indicates that the NMDA receptor subunits, GRIN2A and GRIN2B, might mediate the effects of estrogen in the hippocampal functions, thus suggestive of a translational significance of the finding.


Asunto(s)
Estradiol/fisiología , Hipocampo/fisiología , Memoria/fisiología , Trastornos del Humor/fisiopatología , Estrés Psicológico/fisiopatología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Estradiol/farmacología , Femenino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Trastornos del Humor/complicaciones , Ovariectomía , Progesterona/farmacología , Progesterona/fisiología , Receptores de N-Metil-D-Aspartato/biosíntesis , Estrés Psicológico/complicaciones
19.
Biomed Res Int ; 2016: 5843981, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27478831

RESUMEN

Background. Intrauterine hypoxia is a common cause of fetal growth and lung development restriction. Although N-methyl-D-aspartate receptors (NMDARs) are distributed in the postnatal lung and play a role in lung injury, little is known about NMDAR's expression and role in fetal lung development. Methods. Real-time PCR and western blotting analysis were performed to detect NMDARs between embryonic days (E) 15.5 and E21.5 in fetal rat lungs. NMDAR antagonist MK-801's influence on intrauterine hypoxia-induced retardation of fetal lung development was tested in vivo, and NMDA's direct effect on fetal lung development was observed using fetal lung organ culture in vitro. Results. All seven NMDARs are expressed in fetal rat lungs. Intrauterine hypoxia upregulated NMDARs expression in fetal lungs and decreased fetal body weight, lung weight, lung-weight-to-body-weight ratio, and radial alveolar count, whereas MK-801 alleviated this damage in vivo. In vitro experiments showed that NMDA decreased saccular circumference and area per unit and downregulated thyroid transcription factor-1 and surfactant protein-C mRNA expression. Conclusions. The excessive activation of NMDARs contributed to hypoxia-induced fetal lung development retardation and appropriate blockade of NMDAR might be a novel therapeutic strategy for minimizing the negative outcomes of prenatal hypoxia on lung development.


Asunto(s)
Desarrollo Fetal/genética , Lesión Pulmonar/genética , Pulmón/crecimiento & desarrollo , Receptores de N-Metil-D-Aspartato/genética , Animales , Hipoxia de la Célula/genética , Maleato de Dizocilpina/administración & dosificación , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/patología , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Técnicas de Cultivo de Órganos , ARN Mensajero/biosíntesis , Ratas , Receptores de N-Metil-D-Aspartato/biosíntesis , Factor Nuclear Tiroideo 1 , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
20.
Clin Exp Pharmacol Physiol ; 43(10): 967-75, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27333430

RESUMEN

Hyperhomocysteine has become a critical risk for atherosclerosis and can stimulate proliferation and migration of vascular smooth muscle cells (VSMCs). N-methyl-D-aspartic acid receptor (NMDAR) is a receptor of homocysteine and mediates the effects of homocysteine on VSMCs. Bioinformatics analysis has shown NMDAR is a potential target of microRNA-217 (miR-217), which exerts multiple functions in cancer tumorigenesis and carotid plaque progression. In this study, we sought to investigate the role of miR-217 in VSMCs phenotype transition under homocysteine exposure and elucidate its effect on atherosclerotic plaque formation. After treating with several doses of homocysteine (0-8 × 10(-4)  mol/L) for 24 hours, the expression of miR-217 in HA-VSMCs and rat aortic VSMCs was not altered. Intriguingly, the expression of NMDAR mRNA and protein was reduced by homocysteine in a dose-dependent manner. Transfection of miR-217 mimic significantly inhibited the proliferation and migration of VSMCs with homocysteine treatment, while transfection of miR-217 inhibitor promoted VSMCs migration. Moreover, miR-217 mimic down-regulated while miR-217 inhibitor up-regulated NMDAR protein expression but not NMDAR mRNA expression. Through luciferase reporter assay, we showed that miR-217 could directly bind to the 3'-UTR of NMDAR. MiR-217 mimic transfection also released the inhibition of cAMP-response element-binding protein (CREB)-PGC-1α signalling induced by homocysteine. Additionally, restoration of PGC-1α expression via AdPGC-1α infection markedly suppressed VSMCs proliferation through the degradation of NADPH oxidase (NOX1) and reduction of reactive oxygen species (ROS). Collectively, our study identified the role of miR-217 in regulating VSMCs proliferation and migration, which might serve as a target for atherosclerosis therapy.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular/fisiología , Homocisteína/farmacología , MicroARNs/biosíntesis , Músculo Liso Vascular/metabolismo , Receptores de N-Metil-D-Aspartato/biosíntesis , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Músculo Liso Vascular/efectos de los fármacos , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA