Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(16)2021 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-34445210

RESUMEN

Ischemic episodes are a leading cause of death worldwide with limited therapeutic interventions. The current study explored mitochondrial phosphate-activated glutaminase (GLS1) activity modulation by PKCßII through GC-MS untargeted metabolomics approach. Mitochondria were used to elucidate the endogenous resistance of hippocampal CA2-4 and dentate gyrus (DG) to transient ischemia and reperfusion in a model of ischemic episode in gerbils. In the present investigation, male gerbils were subjected to bilateral carotids occlusion for 5 min followed by reperfusion (IR). Gerbils were randomly divided into three groups as vehicle-treated sham control, vehicle-treated IR and PKCßII specific inhibitor peptide ßIIV5-3-treated IR. Vehicle or ßIIV5-3 (3 mg/kg, i.v.) were administered at the moment of reperfusion. The gerbils hippocampal tissue were isolated at various time of reperfusion and cell lysates or mitochondria were isolated from CA1 and CA2-4,DG hippocampal regions. Recombinant proteins PKCßII and GLS1 were used in in vitro phosphorylation reaction and organotypic hippocampal cultures (OHC) transiently exposed to NMDA (25 µM) to evaluate the inhibition of GLS1 on neuronal viability. PKCßII co-precipitates with GAC (GLS1 isoform) in CA2-4,DG mitochondria and phosphorylates GLS1 in vitro. Cell death was dose dependently increased when GLS1 was inhibited by BPTA while inhibition of mitochondrial pyruvate carrier (MPC) attenuated cell death in NMDA-challenged OHC. Fumarate and malate were increased after IR 1h in CA2-4,DG and this was reversed by ßIIV5-3 what correlated with GLS1 activity increases and earlier showed elevation of neuronal death (Krupska et al., 2017). The present study illustrates that CA2-4,DG resistance to ischemic episode at least partially rely on glutamine and glutamate utilization in mitochondria as a source of carbon to tricarboxylic acid cycle. This phenomenon depends on modulation of GLS1 activity by PKCßII and remodeling of MPC: all these do not occur in ischemia-vulnerable CA1.


Asunto(s)
Trastornos Cerebrovasculares/enzimología , Glutaminasa/metabolismo , Hipocampo/enzimología , Mitocondrias/enzimología , Proteína Quinasa C beta/metabolismo , Daño por Reperfusión/enzimología , Animales , Trastornos Cerebrovasculares/patología , Gerbillinae , Hipocampo/patología , Mitocondrias/patología , Ratas , Ratas Wistar , Daño por Reperfusión/patología
2.
Nutrients ; 12(7)2020 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-32709137

RESUMEN

BACKGROUND: Acute liver failure (ALF) impairs cerebral function and induces hepatic encephalopathy (HE) due to the accumulation of neurotoxic and neuroactive substances in the brain. Cerebral oxidative stress (OS), under control of the glutathione-based defense system, contributes to the HE pathogenesis. Glutathione synthesis is regulated by cysteine synthesized from homocysteine via the transsulfuration pathway present in the brain. The transsulfuration-transmethylation interdependence is controlled by a methyl group donor, S-adenosylmethionine (AdoMet) conversion to S-adenosylhomocysteine (AdoHcy), whose removal by subsequent hydrolysis to homocysteine counteract AdoHcy accumulation-induced OS and excitotoxicity. METHODS: Rats received three consecutive intraperitoneal injections of thioacetamide (TAA) at 24 h intervals. We measured AdoMet and AdoHcy concentrations by HPLC-FD, glutathione (GSH/GSSG) ratio (Quantification kit). RESULTS: AdoMet/AdoHcy ratio was reduced in the brain but not in the liver. The total glutathione level and GSH/GSSG ratio, decreased in TAA rats, were restored by AdoMet treatment. CONCLUSION: Data indicate that disturbance of redox homeostasis caused by AdoHcy in the TAA rat brain may represent a deleterious mechanism of brain damage in HE. The correction of the GSH/GSSG ratio following AdoMet administration indicates its therapeutic value in maintaining cellular redox potential in the cerebral cortex of ALF rats.


Asunto(s)
Encéfalo/efectos de los fármacos , Fallo Hepático Agudo/metabolismo , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Tioacetamida/toxicidad , Animales , Encéfalo/metabolismo , Cistationina betasintasa/metabolismo , Glutatión/metabolismo , Encefalopatía Hepática/etiología , Encefalopatía Hepática/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Fallo Hepático Agudo/inducido químicamente , Masculino , Metionina Adenosiltransferasa/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
3.
Cytokine ; 123: 154774, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31344597

RESUMEN

Glutamate related excitotoxicity and excess of cerebral levels of tumor necrosis factor alpha (TNFα) are interrelated and well documented abnormalities noticed in many central nervous system diseases. Contribution of kidney type glutaminase (KGA) and shorter alternative splicing form (GAC) to glutamine degradation in astrocytes has been recently a matter of dispute and extensive study but the regulation of the GLS isoforms by inflammatory factors is still not well known. Here we show that treatment of cultured rat cortical astrocytes with pathophysiologically relevant (50 ng/ml) concentration of TNFα specifically increases the expression of KGA but not GAC and increases activity of GLS. No changes in the expression of either of two GLS isoforms were observed following treatment with other tested cytokines IL-1ß and IL-6. The TNFα mediated KGA expression was associated with increased phosphorylation of signal transducer and activator of transcription 3 (STAT3). Stimulatory effect of TNF-α on KGA expression was reduced by selective inhibition of (STAT3) but not by inhibition of STAT1 nor nuclear transcription factor kappa. Additionally, the role of miRNA in TNFα-induced expression of KGA in astrocytes was excluded, since the expression of miR-23a/b and miR-200c, potential regulators of KGA expression, was unchanged. This study documents increased KGA expression in the astrocytes under inflammatory stimulation, identifying TNFα as a cytokine mediating this response, and demonstrates the specific and selective involvement of STAT3.


Asunto(s)
Astrocitos/inmunología , Regulación Enzimológica de la Expresión Génica/inmunología , Glutaminasa/inmunología , Factor de Transcripción STAT3/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Astrocitos/citología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Isoenzimas/inmunología , Ratas , Ratas Wistar
4.
Mol Neurobiol ; 56(1): 125-140, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29681024

RESUMEN

α-Synuclein (ASN) and parkin, a multifunctional E3 ubiquitin ligase, are two proteins that are associated with the pathophysiology of Parkinson's disease (PD). Excessive release of ASN, its oligomerization, aggregation, and deposition in the cytoplasm contribute to neuronal injury and cell death through oxidative-nitrosative stress induction, mitochondrial impairment, and synaptic dysfunction. In contrast, overexpression of parkin provides protection against cellular stresses and prevents dopaminergic neural cell loss in several animal models of PD. However, the influence of ASN on the function of parkin is largely unknown. Therefore, the aim of this study was to investigate the effect of extracellular ASN oligomers on parkin expression, S-nitrosylation, as well as its activity. For these investigations, we used rat pheochromocytoma (PC12) cell line treated with exogenous oligomeric ASN as well as PC12 cells with parkin overexpression and parkin knock-down. The experiments were performed using spectrophotometric, spectrofluorometric, and immunochemical methods. We found that exogenous ASN oligomers induce oxidative/nitrosative stress leading to parkin S-nitrosylation. Moreover, this posttranslational modification induced the elevation of parkin autoubiquitination and degradation of the protein. The decreased parkin levels resulted in significant cell death, whereas parkin overexpression protected against toxicity induced by extracellular ASN oligomers. We conclude that lowering parkin levels by extracellular ASN may significantly contribute to the propagation of neurodegeneration in PD pathology through accumulation of defective proteins as a consequence of parkin degradation.


Asunto(s)
Espacio Extracelular/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Ubiquitina-Proteína Ligasas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Supervivencia Celular , Homeostasis , Humanos , Óxido Nítrico/metabolismo , Nitrosación , Estrés Oxidativo , Células PC12 , Multimerización de Proteína , Ratas , Ubiquitinación , alfa-Sinucleína/química , alfa-Sinucleína/ultraestructura
5.
Front Mol Neurosci ; 11: 418, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30505268

RESUMEN

The disorders of the glutamatergic neurotransmission have been associated with pathogenesis of autism. In this study we evaluated the impact of the in vivo and ex vivo test methodology on measurements of levels of neurotransmitter amino acids in hippocampus of rats for valproic acid- (VPA) and thalidomide- (THAL) induced models of autism. The main goal was to compare the changes in concentrations of glutamate (Glu), glutamine (Gln) and GABA between both autistic groups and the control, measured in vivo and ex vivo in homogenates. The rat pups underwent three in vivo tests: ultrasonic vocalization (USV), magnetic resonance spectroscopy (MRS) and unilateral microdialysis of the hippocampus. Analyses of homogenates of rat hippocampus were performed using high-performance liquid chromatography (HPLC) and nuclear magnetic resonance (NMR) spectroscopy. For the statistical analysis, we performed univariate and multivariate tests. USV test, which is considered in rodents as an indicator of pathology similar to autism, showed decreased USV in VPA and THAL groups. In vivo MRS studies demonstrated increases of Glu content in male rat's hippocampus in VPA and THAL groups, while the microdialysis, which allows examination of the contents in the extracellular space, detected decreases in the basal level of Gln concentrations in VPA and THAL groups. Ex vivo HPLC studies showed that levels of Glu, Gln and GABA significantly increased in male rat's hippocampus in the VPA and THAL groups, while NMR studies showed increased levels of Gln and GABA in the VPA group. Collectively, these results are consistent with the hypothesis suggesting the role of the glutamatergic disturbances on the pathogenesis of autism. For all methods used, the values of measured changes were in the same direction. The orthogonal partial least square discriminant analysis confirmed that both animal models of autism tested here can be used to trace neurochemical changes in the brain.

6.
J Neurochem ; 147(5): 692-704, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30151828

RESUMEN

Cerebral blood flow (CBF) is impaired in acute liver failure (ALF), however, the complexity of the underlying mechanisms has often led to inconclusive interpretations. Regulation of CBF depends at least partially on variations in the local brain L-arginine concentration and/or its metabolic rate. In ALF, other factors, like an increased concentration of asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase inhibitor and elevated level of L-glutamine, may contribute to CBF alteration. This study demonstrated strong differences in the reactivity of the middle cerebral arteries and their response to extravascular L-arginine application between vessels isolated from rats with thioacetamide (TAA)-induced ALF and control animals. Our results also showed the decrease in the cerebral perfusion in TAA rats measured by arterial spin labeling perfusion magnetic resonance. Subsequently, we aimed to investigate the importance of balance between the concentration of ADMA and L-arginine in the CBF regulation. In vivo, intraperitoneal L-arginine administration in TAA rats corrected: (i) decrease in cerebral perfusion, (ii) decrease in brain extracellular L-arginine/ADMA ratio and (iii) increase in brain L-glutamine concentration. Our study implicates that impaired vascular tone of cerebral arteries is most likely associated with exposure to high ADMA and L-glutamine levels resulting in limited availability of L-arginine and might be responsible for reduced cerebral perfusion observed in ALF.


Asunto(s)
Arginina/análogos & derivados , Arginina/farmacología , Circulación Cerebrovascular/efectos de los fármacos , Glutamina/metabolismo , Fallo Hepático Agudo/fisiopatología , Animales , Arginina/metabolismo , Encéfalo/diagnóstico por imagen , Espacio Extracelular/metabolismo , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Arteria Cerebral Media/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Tioacetamida
7.
J Alzheimers Dis ; 62(1): 279-291, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29439324

RESUMEN

Urea cycle enzymes may play important yet poorly characterized roles in Alzheimer's disease (AD). Our previous results showed that amyloid-ß (Aß) affects urea cycle enzymes in rat pheochromocytoma (PC12) cells. The aim of the present study was to investigate the changes in arginases, other urea cycle enzymes, and nitric oxide synthases (NOSs) in PC12 cells transfected with AßPP bearing the double 'Swedish' mutation (APPsw, K670M/N671L) and in postmortem sporadic AD brain hippocampus; the mutation intensifies Aß production and strongly associates with AD neuropathology. mRNA expression was analyzed using real-time PCR in cell cultures and DNA microarrays in hippocampal CA1 area of human AD brains. Arginase activity was measured spectrophotometrically, and arginine, ornithine, and citrulline levels by high-performance liquid chromatography. Our data demonstrated that the expression and activity of arginases (Arg1 and Arg2), as well as the expression of argininosuccinate synthase (Ass) were significantly reduced in APPsw cells compared to control. However, argininosuccinate lyase (Asl) was upregulated in APPsw cells. Real-time PCR analysis revealed significant elevation of neuronal nitric oxide synthase (Nnos) mRNA in APPsw cells, without changes in the endothelial Enos, whereas inducible Inos was undetectable. The changes were found to follow closely those observed in the human hippocampal CA1 region of sporadic AD brains. The changes in enzyme expression were accompanied in APPsw cells by significantly elevated citrulline, ornithine, and arginine. Our findings demonstrate that AßPP/Aß alters arginine metabolism and induces a shift of cellular homeostasis that may support the oxidative/nitrosative stress observed in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Arginasa/metabolismo , Región CA1 Hipocampal/metabolismo , Óxido Nítrico Sintasa/metabolismo , Urea/metabolismo , Enfermedad de Alzheimer/patología , Animales , Arginina/metabolismo , Argininosuccinatoliasa/metabolismo , Argininosuccinato Sintasa/metabolismo , Región CA1 Hipocampal/patología , Regulación de la Expresión Génica , Homeostasis/fisiología , Humanos , Células PC12 , ARN Mensajero/metabolismo , Ratas
8.
Int J Mol Sci ; 18(11)2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29099056

RESUMEN

Previously we had shown that ammonia stimulates nitric oxide (NO) synthesis in astrocytes by increasing the uptake of the precursor amino acid, arginine via the heteromeric arginine/glutamine transporter y⁺LAT2. Ammonia also increases the concentration in the brain of the endogenous inhibitor of nitric oxide synthases (NOS), asymmetric dimethylarginine (ADMA), but distribution of ADMA surplus between the intraastrocytic and extracellular compartments of the brain has not been studied. Here we tested the hypothesis that ammonia modulates the distribution of ADMA and its analog symmetric dimethylarginine (SDMA) between the two compartments of the brain by competition with arginine for the y⁺LAT2 transporter. In extension of the hypothesis we analyzed the ADMA/Arg interaction in endothelial cells forming the blood-brain barrier. We measured by high-performance liquid chromatography (HPLC) and mass spectrometry (MS) technique the concentration of arginine, ADMA and SDMA in cultured cortical astrocytes and in a rat brain endothelial cell line (RBE-4) treated with ammonia and the effect of silencing the expression of a gene coding y⁺LAT2. We also tested the expression of ADMA metabolism enzymes: protein arginine methyltransferase (PRMT) and dimethylarginine dimethyl aminohydrolase (DDAH) and arginine uptake to astrocytes. Treatment for 48 h with 5 mM ammonia led to an almost 50% reduction of ADMA and SDMA concentration in both cell types, and the effect in astrocytes was substantially attenuated by silencing of the Slc7a6 gene. Moreover, the y⁺LAT2-dependent component of ammonia-evoked arginine uptake in astrocytes was reduced in the presence of ADMA in the medium. Our results suggest that increased ADMA efflux mediated by upregulated y⁺LAT2 may be a mechanism by which ammonia interferes with intra-astrocytic (and possibly intra-endothelial cell) ADMA content and subsequently, NO synthesis in both cell types.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Amoníaco/metabolismo , Arginina/análogos & derivados , Astrocitos/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Amidohidrolasas/metabolismo , Animales , Arginina/metabolismo , Línea Celular , Células Cultivadas , Óxido Nítrico/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Ratas , Ratas Wistar
9.
Curr Alzheimer Res ; 13(9): 1030-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26971935

RESUMEN

Alterations of enzymes linked to arginine metabolism have been recently implicated in Alzheimer's disease (AD). Despite strong association of arginine changes with nitric oxide (NO) pathway, the impact of amyloid ß (Aß) peptides on arginine degradation and re-synthesis is unknown. In the present study we compared expression levels of arginases (ARG1, ARG2), neuronal, endothelial and inducible NO synthase isoforms (NNOS, ENOS, INOS), enzymes that metabolize arginine or resynthesize it from citrulline and the levels of corresponding amino acids in rat pheochromocytoma (PC12) cells overexpressing human Aß precursor protein (APPwt cells). Moreover, we investigated the changes in miRNAs responsible for modulation of arginine metabolism in AD brains. Real-time PCR analysis revealed in APPwt cells significant decreases of ARG1 and ARG2 which are responsible for lysing arginine into ornithine and urea; this reduction was followed by significantly lower enzyme activity. NNOS and ENOS mRNAs were elevated in APPwt cells while iNOS was undetectable in both cell lines. The expression of argininosuccinate synthase (ASS) that metabolizes citrulline was down-regulated without changes in argininosuccinate lyase (ASL). Ornithine decarboxylase (ODC), which decarboxylates ornithine to form putrescine was also reduced. Arginine, the substrate for both arginases and NOS, was unchanged in APPwt cells. However, citrulline concentration was significantly higher. Elevated miRNA-9 and miRNA-128a found in AD brain tissues might modulate the expression of ASS and NOS, respectively. Our results indicate that Aß affects arginine metabolism and this influence might have important role in the pathomechanism of AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Arginina/metabolismo , Anciano , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Arginasa/metabolismo , Encéfalo/metabolismo , Humanos , MicroARNs/metabolismo , Óxido Nítrico Sintasa/metabolismo , Células PC12 , ARN Mensajero/metabolismo , Ratas , Transfección
10.
Neurochem Res ; 41(1-2): 376-84, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26801175

RESUMEN

Cerebral oxidative stress (OS) contributes to the pathogenesis of hepatic encephalopathy (HE). Existing evidence suggests that systemic administration of L-histidine (His) attenuates OS in brain of HE animal models, but the underlying mechanism is complex and not sufficiently understood. Here we tested the hypothesis that dipeptide carnosine (ß-alanyl-L-histidine, Car) may be neuroprotective in thioacetamide (TAA)-induced liver failure in rats and that, being His metabolite, may mediate the well documented anti-OS activity of His. Amino acids [His or Car (100 mg/kg)] were administrated 2 h before TAA (i.p., 300 mg/kg 3× in 24 h intervals) injection into Sprague-Dawley rats. The animals were thus tested for: (i) brain prefrontal cortex and blood contents of Car and His, (ii) amount of reactive oxygen species (ROS), total antioxidant capacity (TAC), GSSG/GSH ratio and thioredoxin reductase (TRx) activity, and (iii) behavioral changes (several models were used, i.e. tests for reflexes, open field, grip test, Rotarod). Brain level of Car was reduced in TAA rats, and His administration significantly elevated Car levels in control and TAA rats. Car partly attenuated TAA-induced ROS production and reduced GSH/GSSG ratio, whereas the increase of TRx activity in TAA brain was not significantly modulated by Car. Further, Car improved TAA-affected behavioral functions in rats, as was shown by the tests of righting and postural reflexes. Collectively, the results support the hypothesis that (i) Car may be added to the list of neuroprotective compounds of therapeutic potential on HE and that (ii) Car mediates at least a portion of the OS-attenuating activity of His in the setting of TAA-induced liver failure.


Asunto(s)
Carnosina/farmacología , Fallo Hepático/inducido químicamente , Estrés Oxidativo/efectos de los fármacos , Postura , Tioacetamida/toxicidad , Animales , Fallo Hepático/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley
11.
Folia Neuropathol ; 53(4): 377-86, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26785372

RESUMEN

INTRODUCTION: Our previous studies showed only slight improvement in central fatigue, measured indirectly by psychomotor performance, after branched chain amino acids (BCAA) supplementation during various efforts in healthy men. It is hypothesised that hyperammonaemia resulting from amino acids metabolism may attenuate their beneficial effect on psychomotor performance; therefore, the L-ornithine L-aspartate (OA) as an ammonia decreasing agent was used. The aim of this study was to investigate the effectiveness of oral BCAA + OA supplementation to reduce plasma ammonia concentration and enhance psychomotor performance during exhaustive exercise in healthy men. MATERIAL AND METHODS: Eleven endurance-trained men (mean age 32.6 ± 1.9 years) performed two sessions (separated by one week) of submaximal cycloergometer exercise for 90 minutes at 60% of maximal oxygen uptake followed by graded exercise until exhaustion with randomised, double-blind supplementation with a total of 16 g BCAA and 12 g OA (BCAA + OA trial) or flavoured water (placebo trial). Before exercise, during both efforts and after 20 minutes of recovery multiple choice reaction time (MCRT), perceived exertion, heart rate and oxygen uptake were measured and venous blood samples were taken for plasma leucine, valine, isoleucine, ornithine, aspartate, free tryptophan (fTRP), ammonia, lactate and glucose determination. RESULTS: After ingestion, during both efforts and after 20 minutes of recovery the plasma concentrations of all supplemented amino acids were significantly increased, while the fTRP/BCAA ratio decreased in the BCAA + OA trial more than in the placebo trial. At the end of graded exercise plasma fTRP was lower and MCRT shorter in BCAA + OA than in the placebo trial (p < 0.05). At the end of prolonged exercise the plasma ammonia concentration was higher in BCAA + OA than in placebo trial (p < 0.05). Decreases in plasma ammonia during recovery were significantly higher in BCAA + OA than in the placebo trial. Plasma ammonia positively correlated with the total plasma BCAA and MCRT only in the BCAA + OA trial. The fTRP/BCAA ratio positively correlated with MCRT only in the placebo trial. CONCLUSIONS: Supplementation with BCAA and OA is a useful way to improve MCRT during high-intensity exercise and accelerate the elimination of ammonia at the recovery stage after exercise in healthy young men.


Asunto(s)
Aminoácidos de Cadena Ramificada/administración & dosificación , Amoníaco/sangre , Suplementos Dietéticos , Dipéptidos/administración & dosificación , Ejercicio Físico , Fatiga Muscular/efectos de los fármacos , Adulto , Estudios Cruzados , Método Doble Ciego , Ejercicio Físico/fisiología , Estado de Salud , Humanos , Masculino , Fatiga Muscular/fisiología , Resistencia Física/efectos de los fármacos , Resistencia Física/fisiología
12.
Neurochem Int ; 88: 26-31, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25523831

RESUMEN

Hepatic encephalopathy (HE) is related to variations in the nitric oxide (NO) synthesis and oxidative/nitrosative stress (ONS), and asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthases (NOSs). In the present study we compared the effects of acute liver failure (ALF) in the rat TAA model on ADMA concentration in plasma and cerebral cortex, and on the activity and expression of the ADMA degrading enzyme, dimethylarginine dimethylaminohydrolase (DDAH), in brain and liver. ALF increased blood and brain ADMA, and the increase was correlated with decreased DDAH activity in both brain and liver. An i.p. administration of histidine (His), an amino acid reported to alleviate oxidative stress associated with HE (100 mg/kg b.w.), reversed the increase of brain ADMA, which was accompanied by the recovery of brain DDAH activity (determined ex vivo), and with an increase of the total NOS activity. His also activated DDAH ex vivo in brain homogenates derived from control and TAA rats. ALF in this model was also accompanied by increases of blood cyclooxygenase activity and blood and brain TNF-α content, markers of the inflammatory response in the periphery, but these changes were not affected by His, except for the reduction of TNF-α mRNA transcript in the brain. His increased the total antioxidant capacity of the brain cortex, but not of the blood, further documenting its direct neuroprotective power.


Asunto(s)
Arginina/análogos & derivados , Encéfalo/metabolismo , Histidina/farmacología , Fallo Hepático Agudo/metabolismo , Óxido Nítrico/metabolismo , Tioacetamida/toxicidad , Animales , Arginina/metabolismo , Encéfalo/efectos de los fármacos , Histidina/fisiología , Fallo Hepático Agudo/inducido químicamente , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
13.
Pharmacol Rep ; 66(3): 466-70, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24905525

RESUMEN

BACKGROUND: Kynurenic acid (KYNA) modulates the glutamatergic tone by controlling neuronal glutamate (GLU) release. The present study tested the potential of the KYNA precursor, kynurenine (KYN) to counter increased extracellular GLU associated with the pathogenesis of hepatic encephalopathy accompanying acute liver failure (ALF). METHODS: ALF was induced in adult rats by administration of a hepatotoxin, thioacetamide. KYNA and GLU were measured in the cerebral cortical microdialysates of control (saline-treated) and ALF rats using HPLC. The expression of mRNA coding for kynurenine aminotransferase II (KAT-II), the astrocytic enzyme converting KYN to KYNA, was assayed by real-time PCR. RESULTS: Cerebral cortical extracellular KYNA was increased in ALF rats not treated with KYN, consistent with a previously observed increase of cerebral cortical KATII activity in this ALF model. Single intraperitoneal administration of KYN (50 mg/kg, 120 min before microdialysate collection), produced a further substantial increase of extracellular KYNA, paralleled by a decrease of extracellular GLU. In cultured cerebral cortical astrocytes, the cells which in situ are the primary target of blood-derived ammonia and other toxins liberated due to ALF, elevation of KAT-II mRNA expression was noted upon their incubation with KYN and the KYN precursor, tryptophan (Trp), which is normally elevated by ALF. CONCLUSIONS: Administration of exogenous KYN to stimulate KYNA synthesis may help correcting excessive extracellular accumulation of GLU in cerebral cortex caused by ALF. The therapeutic potential of KYN in ALF appears to be fostered by increased expression of KAT-II in astrocytes upon exposure to KYN or Trp.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Ácido Glutámico/metabolismo , Ácido Quinurénico/metabolismo , Quinurenina/farmacología , Fallo Hepático Agudo/tratamiento farmacológico , Animales , Corteza Cerebral/metabolismo , Fallo Hepático Agudo/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Transaminasas/metabolismo
14.
Metab Brain Dis ; 29(4): 1053-60, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24385142

RESUMEN

L-citrulline (Cit) is a co-product of NO synthesis and a direct L-arginine (Arg) precursor for de novo NO synthesis. Acute liver failure (ALF) is associated with increased nitric oxide (NO) and cyclic GMP (cGMP) synthesis in the brain, indirectly implicating a role for active transport of Cit. In the present study we characterized [(3)H]Cit uptake to the cortical brain slices obtained from control rats and rats with thioacetamide (TAA)-induced ALF ("TAA slices"). In both control and TAA slices the uptake was partially Na(+)-dependent and markedly inhibited by substrates of systems L and N, including L-glutamine (Gln), which accumulates in excess in brain during ALF. Cit uptake was not affected by Arg, the y(+)/y(+)L transport system substrate, nor by amino acids taken up by systems A, xc (-)or XAG. The Vmax of the uptake in TAA slices was ~60 % higher than in control slices. Chromatographic (HPLC) analysis revealed a ~30 % increase of Cit concentration in the cerebral cortical homogenates of TAA rats. The activity of argininosuccinate synthase (ASS) and argininosuccinate lyase (ASL), the two enzymes of Cit-NO cycle catalyzing synthesis of Arg, showed an increase in TAA rats, consistent with increased ASS and ASL protein expression, by ~30 and ~20 %, respectively. The increased Cit-NO cycle activity was paralleled by increased expression of mRNA coding for inducible nitric oxide synthase (iNOS). Taken together, the results suggest a role for Cit in the activation of cerebral NO synthesis during ALF.


Asunto(s)
Corteza Cerebral/metabolismo , Citrulina/metabolismo , Fallo Hepático/inducido químicamente , Tioacetamida/toxicidad , Animales , Arginina/metabolismo , Argininosuccinatoliasa/metabolismo , Argininosuccinato Sintasa/metabolismo , Técnicas In Vitro , Cinética , Fallo Hepático/metabolismo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Óxido Nítrico/biosíntesis , Ratas , Ratas Sprague-Dawley
15.
Tumour Biol ; 35(3): 1855-62, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24096582

RESUMEN

Glutamine (Gln) metabolism, initiated by its degradation by glutaminases (GA), is elevated in neoplastic cells and tissues. In malignant glia-derived tumors, GA isoforms, KGA and GAC, coded by the GLS gene, are overexpressed, whereas the GLS2-coded GAB and LGA isoforms, are hardly detectable in there. Our previous study revealed that transfection of T98G glioblastoma cells with GAB reduced cell proliferation and migration, by a yet unknown mechanism not related to Gln degradation. The question arose how simultaneous overexpression of GAB and inhibition of KGA would affect glioblastoma cell growth. Here, we used siRNA to silence the expression of Gls in T98G cells which were or were not stably transfected with GAB (TGAB cells). In both T98G and TGAB cell lines, silencing of Gls with siRNAs targeted at different sequences decreased cell viability and proliferation in a different, sequence-dependent degree, and the observed decreases were in either cell line highly correlated with increase of intracellular Gln (r > 0.9), a parameter manifesting decreased Gln degradation. The results show that combination of negative modulation of GA isoforms arising from GLS gene with the introduction of the GLS2 gene product, GAB, may in the future provide a useful means to curb glioblastoma growth in situ. At the same time, the results underscore the critical role of Gln degradation mediated by KGA in the manifestations of aggressive glial tumor phenotype.


Asunto(s)
Proliferación Celular , Glioblastoma/enzimología , Glioblastoma/genética , Glutaminasa/genética , Glutaminasa/metabolismo , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/genética , Silenciador del Gen , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
16.
Metab Brain Dis ; 28(1): 11-4, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23086200

RESUMEN

Mitochondrial glutathione (mGSH) is a critical factor in the cell defense against oxidative and nitrosative stress (ONS), and ONS is a key pathogenic event in hepatic encephalopathy (HE). Acute HE in the thioacetamide (TAA) model caused a 54 % decrease of mGSH content in the rat prefrontal cortex (pfc), but not in the striatum (str), nor did it affect the GSH content in the pfc or str homogenate. In the pfc, treatment with L- histidine (His), which is known to alleviate ONS-related symptoms in HE animals, attenuated the decrease of mGSH, and increased the GSH content in pfc and str homogenates and pfc microdialysates of control animals. His increased the expression of mRNA coding for the GSH synthesizing enzyme, glutamate cysteine ligase (GCL) and decreased that of the GSH-degrading enzyme γ-glutamyltranspeptidase (γGT). The results suggest that the decrease of mGSH may be an important contributing factor to mitochondrial dysfunction in HE, and delineate a new mechanistic aspect of the therapeutic potential of His in HE.


Asunto(s)
Glutatión/análisis , Encefalopatía Hepática/metabolismo , Histidina/uso terapéutico , Mitocondrias/metabolismo , Corteza Prefrontal/metabolismo , Enfermedad Aguda , Animales , Glutamato-Cisteína Ligasa/genética , Glutatión/metabolismo , Encefalopatía Hepática/tratamiento farmacológico , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
17.
Neurotoxicology ; 33(1): 16-22, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22044768

RESUMEN

Aroclor 1254 is a mixture of polychlorinated biphenyls (PCBs), a class of environmental toxins which cause a wide spectrum of neurotoxic effects. Learning and memory deficits are the profound effects of PCBs which may be related to hippocampal dysfunction. To get insight into the underlying neurochemical mechanisms, we employed the microdialysis technique to investigate the effect of repeated exposure of adult male Wistar rats to Aroclor 1254 (10mg/kg b.w., daily, ig., for 14days), on the neurochemical parameters of NMDA receptor-mediated glutamatergic signaling in the hippocampus in vivo assessed using the microdialysis technique. The results demonstrated that exposure to Aroclor 1254, which was associated with substantial neuronal damage and loss in the hippocampus, markedly decreased the NMDA-induced extracellular accumulation of newly loaded (45)CaCl(2), cGMP and glutamate, and reduced the basal content of the NO precursor, arginine, indicating inhibition of the NMDA/NO/cGMP pathway. Aroclor 1254 exposure also decreased the basal microdialysate content of glutamate and glutamine, which may cause inadequate supply of the neurotransmitter glutamate, while the level of two other neuroactive amino acids, aspartate or taurine was not affected by the exposure. The results underscore neuronal lesion and inhibition of NMDA receptor-mediated glutamatergic signaling in hippocampus as a potential major contributor to the cognitive deficits associated with exposure to PCB.


Asunto(s)
/farmacología , Contaminantes Ambientales/farmacología , Hipocampo/citología , Neuronas/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Aminoácidos/metabolismo , Animales , Isótopos de Calcio/metabolismo , GMP Cíclico/metabolismo , Agonistas de Aminoácidos Excitadores/farmacología , Hipocampo/lesiones , Masculino , Microdiálisis , Microscopía Electrónica de Transmisión , N-Metilaspartato/farmacología , Neuronas/metabolismo , Neuronas/ultraestructura , Ratas , Ratas Wistar
18.
J Neurosci Res ; 89(12): 2018-27, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21538466

RESUMEN

Brain edema in acute hepatic encephalopathy (HE) is due mainly to swelling of astrocytes. Efflux of potassium is implicated in the prevention of glial swelling under hypoosmotic conditions. We investigated whether pathogenic factors of HE, glutamine (Gln) and/or ammonia, induce alterations in the expression of glial potassium channels (Kir4.1, Kir2.1) and Na(+) -K(+) -2Cl(-) cotransporter-1 (NKCC1) in rat cerebral cortex and cultured rat cortical astrocytes and whether these alterations have consequences for potassium efflux and astrocytic swelling. Thioacetamide-induced acute liver failure in rats resulted in significant decreases in the Kir4.1 mRNA and protein contents of cerebral cortex, whereas expression of Kir2.1 and NKCC1 remained unaltered. Incubation of primary cortical astrocytes for 72 hr in the presence of Gln (5 mM), but not of ammonia (5 mM or 10 mM), induced a decrease in the levels of Kir4.1 mRNA and protein. Similarly to incubation with Gln, reduction of Kir4.1 mRNA expression by RNA interference caused swelling of astrocytes as shown by confocal imaging followed by 3D computational analysis. Gln reduced the astrocytic uptake of D-[(3) H]aspartate, but, in contrast to the earlier reported effect of ammonia, this reduction was not accompanied by decreased expression of the astrocytic glutamate transporter GLT-1 mRNA. Both Gln and ammonia decreased hypoosmolarity-induced (86) Rb efflux from the cells, but the effect was more pronounced with Gln. The results indicate that down-regulation of Kir4.1 may mediate distinct aspects of Gln-induced astrocytic dysfunction in HE.


Asunto(s)
Astrocitos/metabolismo , Encefalopatía Hepática/metabolismo , Fallo Hepático/metabolismo , Canales de Potasio de Rectificación Interna/biosíntesis , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Western Blotting , Células Cultivadas , Corteza Cerebral/metabolismo , Regulación hacia Abajo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Glutamina/farmacología , Encefalopatía Hepática/patología , Masculino , Ratas , Ratas Long-Evans , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Simportadores de Cloruro de Sodio-Potasio/biosíntesis , Miembro 2 de la Familia de Transportadores de Soluto 12
19.
Neurochem Int ; 58(2): 190-5, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21115085

RESUMEN

The pathogenesis of hepatic encephalopathy (HE) is associated with hyperammonemia (HA) and subsequent exposure of the brain to excess of ammonia. Alterations of the NO/cGMP pathway and increased glutamine (Gln) content are collectively responsible for many HE symptoms, but how the two events influence each other is not clear. Previously we had shown that Gln administered intracerebrally inhibited the NO/cGMP pathway in control rats and even more so in rats with HA, and we speculated that this effect is due to inhibition by Gln of arginine (Arg) transport (Hilgier et al., 2009). In this study we demonstrate that a 3-day HA in the ammonium acetate model increases the expression in the brain of y(+)LAT2, the heteromeric transporter which preferentially stimulates Arg efflux from the cells in exchange for Gln. The expression of the basic amino acid transporter CAT1, transporting Arg but not Gln remained unaffected by HA. Multiple parameters of Arg or Gln uptake and/or efflux and their mutual dependence were altered in the cerebral cortical slices obtained from HA rats, in a manner indicating enhanced y(+)LAT2-mediated transport. HA elevated Gln content and decreased cGMP content as measured both in the cerebral cortical tissue and microdialysates. Intracortical administration of 6-diazo-5-oxo-L-norleucine (DON), which inhibits Gln fluxes between different cells of the CNS, attenuated the HA-induced decrease of cGMP in the microdialysates of HA rats, but not of control rats. The results suggest that, reduced delivery of Arg due to enhanced y(+)LAT2-mediated exchange of extracellular Gln for intracellular Arg may contribute to the decrease of NO/cGMP pathway activity evoked in the brain by HA.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Corteza Cerebral/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Encefalopatía Hepática/metabolismo , Hiperamonemia/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal/fisiología , Sistema de Transporte de Aminoácidos y+/genética , Animales , Corteza Cerebral/fisiopatología , GMP Cíclico/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/genética , Encefalopatía Hepática/fisiopatología , Hiperamonemia/fisiopatología , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Ratas Wistar
20.
Toxicol Sci ; 117(1): 163-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20530233

RESUMEN

We endeavored here to shed light on the supply of glutathione (GSH) precursors from glial cells to neurons and on the interference of ammonia with this process. Administration of ammonium chloride (ammonia) via a microdialysis probe to the rat prefrontal cortex rapidly increased GSH content in the microdialysates. The increase was abrogated by the inhibitor of astrocytic energy metabolism fluoroacetate and the inhibitor of glutathione synthesis buthionine sulfoximine. GSH in the microdialysates was significantly elevated in rats with simple hyperammonemia (HA) or hepatic encephalopathy (HE) (three ip administrations of ammonium acetate or thioacetamide, respectively, at 24-h intervals), only when microdialysis was carried out in the presence of a gamma-glutamyltranspeptidase (gammaGT) inhibitor acivicin. Extracellular GSH increased in cultured rat cortical astrocytes treated with 5mM ammonia for 1 h, but not for 3-72 h, which was the period of increased gammaGT activity. GSH remained increased during the whole 72-h incubation with 5 or 10mM ammonia in C6 glioma cells, where gammaGT activity is intrinsically low and was not increased by ammonia. Collectively, the results suggest that in rats with HA or HE ammonia specifically promote GSH synthesis and export from astrocytes and increase its extracellular degradation, which may improve the availability of precursors for GSH synthesis in neurons and their resistance to ammonia toxicity.


Asunto(s)
Amoníaco/toxicidad , Astrocitos/efectos de los fármacos , Glutatión/metabolismo , Hiperamonemia/metabolismo , Corteza Prefrontal/efectos de los fármacos , Amoníaco/sangre , Animales , Astrocitos/metabolismo , Línea Celular Tumoral , Espacio Extracelular/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA