Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Inflamm Res ; 72(2): 203-215, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36401631

RESUMEN

OBJECTIVE: This study aimed to investigate the effects of FK506 on experimental sepsis immunopathology. It investigated the effect of FK506 on leukocyte recruitment to the site of infection, systemic cytokine production, and organ injury in mice with sepsis. METHODS: Using a murine cecal ligation and puncture (CLP) peritonitis model, the experiments were performed with wild-type (WT) mice and mice deficient in the gene Nfat1 (Nfat1-/-) in the C57BL/6 background. Animals were treated with 2.0 mg/kg of FK506, subcutaneously, 1 h before the sepsis model, twice a day (12 h/12 h). The number of bacteria colony forming units (CFU) was manually counted. The number of neutrophils in the lungs was estimated by the myeloperoxidase (MPO) assay. The expression of CXCR2 in neutrophils was determined using flow cytometry analysis. The expression of inflammatory cytokines in macrophage was determined using ELISA. The direct effect of FK506 on CXCR2 internalization was evaluated using HEK-293T cells after CXCL2 stimulation by the BRET method. RESULTS: FK506 treatment potentiated the failure of neutrophil migration into the peritoneal cavity, resulting in bacteremia and an exacerbated systemic inflammatory response, which led to higher organ damage and mortality rates. Failed neutrophil migration was associated with elevated CXCL2 chemokine plasma levels and lower expression of the CXCR2 receptor on circulating neutrophils compared with non-treated CLP-induced septic mice. FK506 did not directly affect CXCL2-induced CXCR2 internalization by transfected HEK-293 cells or mice neutrophils, despite increasing CXCL2 release by LPS-treated macrophages. Finally, the CLP-induced response of Nfat1-/- mice was similar to those observed in the Nfat1+/+ genotype, suggesting that the FK506 effect is not dependent on the NFAT1 pathway. CONCLUSION: Our data indicate that the increased susceptibility to infection of FK506-treated mice is associated with failed neutrophil migration due to the reduced membrane availability of CXCR2 receptors in response to exacerbated levels of circulating CXCL2.


Asunto(s)
Neutrófilos , Sepsis , Humanos , Ratones , Animales , Tacrolimus/farmacología , Tacrolimus/uso terapéutico , Células HEK293 , Ratones Endogámicos C57BL , Sepsis/metabolismo , Infiltración Neutrófila
2.
Neurochem Res ; 46(1): 120-130, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32285377

RESUMEN

Most pharmacological studies concerning the beneficial effects of organoselenium compounds have focused on their ability to mimic glutathione peroxidase (GPx). However, mechanisms other than GPx-like activity might be involved on their biological effects. This study was aimed to investigate and compare the protective effects of two well known [(PhSe)2 and PhSeZnCl] and two newly developed (MRK Picolyl and MRK Ester) organoselenium compounds against oxidative challenge in cultured neuronal HT22 cells. The thiol peroxidase and oxidase activities were performed using the glutathione reductase (GR)-coupled assay. In order to evaluate protective effects of the organoselenium compounds against oxidative challenge in neuronal HT22 cells, experiments based on glutamate-induced oxytosis and SIN-1-mediated peroxynitrite generation were performed. The thiol peroxidase activities of the studied organoselenium compounds were smaller than bovine erythrocytes GPx enzyme. Besides, (PhSe)2 and PhSeZnCl showed higher thiol peroxidase and lower thiol oxidase activities compared to the new compounds. MRK Picolyl and MRK Ester, which showed lower thiol peroxidase activity, showed higher thiol oxidase activity. Both pre- or co-treatment with (PhSe)2, PhSeZnCl, MRK Picolyl and MRK Ester protected HT22 cells against glutamate-induced cytotoxicity. (PhSe)2 and MRK Picolyl significantly prevented peroxinitrite-induced dihydrorhodamine oxidation, but this effect was observed only when HT22 were pre-treated with these compounds. The treatment with (PhSe)2 increased the protein expression of antioxidant defences (Prx3, CAT and GCLC) in HT22 cells. Taking together, our results suggest that the biological effects elicited by these compounds are not directly related to their GPx-mimetic and thiol oxidase activities, but might be linked to the up-regulation of endogenous antioxidant defences trough their thiol-modifier effects.


Asunto(s)
Antioxidantes/farmacología , Neuronas/efectos de los fármacos , Compuestos de Organoselenio/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Catalasa/metabolismo , Bovinos , Línea Celular , Glutamato-Cisteína Ligasa/metabolismo , Glutatión Peroxidasa/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones
3.
Mol Med ; 21: 219-26, 2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25822797

RESUMEN

Sepsis-associated encephalopathy (SAE) is associated with an increased rate of morbidity and mortality. It is not understood what the exact mechanism is for the brain dysfunction that occurs in septic patients, but brain inflammation and oxidative stress are a possible theory. Such events can occur through the alteration of molecules that perpetuate the inflammatory response. Thus, it is possible to postulate that CD40 may be involved in this process. The aim of this work is to evaluate the role of CD40-CD40L pathway activation in brain dysfunction associated with sepsis in an animal model. Microglia activation induces the upregulation of CD40-CD40L, both in vitro and in vivo. The inhibition of microglia activation decreases levels of CD40-CD40L in the brain and decreases brain inflammation, oxidative damage and blood brain barrier dysfunction. Despite this, anti-CD40 treatment does not improve mortality in this model. However, it is able to improve long-term cognitive impairment in sepsis survivors. In conclusion, there is a major involvement of the CD40-CD40L signaling pathway in long-term brain dysfunction in an animal model of sepsis.


Asunto(s)
Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Trastornos del Conocimiento/etiología , Sepsis/complicaciones , Sepsis/metabolismo , Transducción de Señal , Animales , Anticuerpos Monoclonales/farmacología , Biomarcadores , Barrera Hematoencefálica/metabolismo , Antígenos CD40/antagonistas & inhibidores , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/metabolismo , Modelos Animales de Enfermedad , Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Encefalitis/metabolismo , Humanos , Estimación de Kaplan-Meier , Microglía/metabolismo , Sepsis/mortalidad , Regulación hacia Arriba
4.
An Acad Bras Cienc ; 87(2 Suppl): 1487-96, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26312430

RESUMEN

Primaquine and chloroquine are used for the treatment of malaria; evidence from the literature suggests that these drugs may induce oxidative stress. In this study we investigated the effects of primaquine and chloroquine on oxidative damage and DNA damage in brain, liver and kidney of rats after 7, 14 and 21 days of administration. Our results demonstrated that primaquine causes DNA damage in brain after 7, 14 and 21 days, and in liver after 7 and 14 days. Moreover, primaquine increases TBARS levels in the kidney and protein carbonyls in the brain after 14 days, and decreases protein carbonyls in the liver after 7 days. Whereas chloroquine causes DNA damage in the kidney after 7 and 14 days, and in the liver after 14 and 21 days, increases TBARS levels in the kidney after 7 days, and decreases TBARS levels in the brain after 21 days. Moreover, decreases protein carbonyls in the liver after 7 and 14 days, and in the brain after 7 and 21 days. However, chloroquine treatment for 14 days increases protein carbonyls in the brain and kidney. In conclusion, these results showed that prolonged treatment with antimalarial may adversely affect the DNA.


Asunto(s)
Antimaláricos/farmacología , Cloroquina/farmacología , Daño del ADN/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Primaquina/farmacología , Animales , Encéfalo/efectos de los fármacos , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Factores de Tiempo
5.
Mol Med ; 18: 1209-19, 2012 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-22735756

RESUMEN

In sepsis, toll-like receptor (TLR)-4 modulates the migration of neutrophils to infectious foci, favoring bacteremia and mortality. In experimental sepsis, organ dysfunction and cytokines released by activated macrophages can be reduced by gastrin-releasing peptide (GRP) receptor (GRPR) antagonist RC-3095. Here we report a link between GRPR and TLR-4 in experimental models and in sepsis patients. RAW 264.7 culture cells were exposed to lipopolysaccharide (LPS) or tumor necrosis factor (TNF)-α and RC-3095 (10 ng/mL). Male Wistar rats were subjected to cecal ligation and puncture (CLP), and RC-3095 was administered (3 mg/kg, subcutaneously); after 6 h, we removed the blood, bronchoalveolar lavage, peritoneal lavage and lung. Human patients with a clinical diagnosis of sepsis received a continuous infusion with RC-3095 (3 mg/kg, intravenous) over a period of 12 h, and plasma was collected before and after RC-3095 administration and, in a different set of patients with systemic inflammatory response syndrome (SIRS) or sepsis, GRP plasma levels were determined. RC-3095 inhibited TLR-4, extracellular-signal-related kinase (ERK)-1/2, Jun NH(2)-terminal kinase (JNK) and Akt and decreased activation of activator protein 1 (AP-1), nuclear factor (NF)-κB and interleukin (IL)-6 in macrophages stimulated by LPS. It also decreased IL-6 release from macrophages stimulated by TNF-α. RC-3095 treatment in CLP rats decreased lung TLR-4, reduced the migration of cells to the lung and reduced systemic cytokines and bacterial dissemination. Patients with sepsis and systemic inflammatory response syndrome have elevated plasma levels of GRP, which associates with clinical outcome in the sepsis patients. These findings highlight the role of GRPR signaling in sepsis outcome and the beneficial action of GRPR antagonists in controlling the inflammatory response in sepsis through a mechanism involving at least inhibition of TLR-4 signaling.


Asunto(s)
Receptores de Bombesina/antagonistas & inhibidores , Sepsis/prevención & control , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Adulto , Animales , Bombesina/administración & dosificación , Bombesina/análogos & derivados , Bombesina/farmacología , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Péptido Liberador de Gastrina/sangre , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-6/sangre , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Modelos Biológicos , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores de Bombesina/metabolismo , Sepsis/sangre , Sepsis/metabolismo , Sepsis/microbiología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/genética , Factor de Transcripción ReIA/metabolismo
6.
J Neural Transm (Vienna) ; 119(11): 1267-74, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22350588

RESUMEN

Sepsis is characterized by systemic biochemical alterations including the central nervous system in the early times and cognitive impairment at later times after sepsis induction in the animal model. Recent studies have shown that, besides its hematological activity, erythropoietin (EPO) has cytoprotective effects on various cells and tissues. In order to corroborate elucidating the effects of alternative drugs for sepsis treatment, we evaluated the effects of both acute and chronic EPO treatment on oxidative stress and energetic metabolism in the hippocampus, and cognitive impairment, respectively, after sepsis induction by cecal ligation and perforation (CLP). To this aim, male Wistar rats underwent CLP with "basic support" or sham operation. In the acute treatment, EPO was administered once immediately after CLP induction. The rats were then killed after 6 and 24 h, and the hippocampus was removed for analysis of oxidative stress and energetic metabolism, respectively. Regarding the chronic treatment, EPO was administered once daily until the 4th day after induction. Aversive memory was tested on the 10th day after surgery. It was observed that the acute use of EPO (a single dose) alters the oxidative parameters and energetic metabolism. Chronic use (4 days) reversed cognitive impairment in the sepsis animal model. Mortality rates were attenuated only during chronic treatment.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Metabolismo Energético/efectos de los fármacos , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Sepsis/metabolismo , Análisis de Varianza , Animales , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Citrato (si)-Sintasa/metabolismo , Trastornos del Conocimiento/etiología , Creatina Quinasa/metabolismo , Modelos Animales de Enfermedad , Transporte de Electrón/efectos de los fármacos , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Inhibición Psicológica , Ligadura/efectos adversos , Masculino , Ratas , Ratas Wistar , Sepsis/complicaciones , Sepsis/etiología , Estadísticas no Paramétricas , Factores de Tiempo
7.
Redox Biol ; 20: 118-129, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30308475

RESUMEN

Oxidative stress and mitochondrial dysfunction are critical events in neurodegenerative diseases; therefore, molecules that increase cellular antioxidant defenses represent a future pharmacologic strategy to counteract such conditions. The aim of this study was to investigate the potential protective effect of (PhSe)2 on mouse hippocampal cell line (HT22) exposed to tert-BuOOH (in vitro model of oxidative stress), as well as to elucidate potential mechanisms underlying this protection. Our results showed that tert-BuOOH caused time- and concentration-dependent cytotoxicity, which was preceded by increased oxidants production and mitochondrial dysfunction. (PhSe)2 pre-incubation significantly prevented these cytotoxic events and the observed protective effects were paralleled by the upregulation of the cellular glutathione-dependent antioxidant system: (PhSe)2 increased GSH levels (> 60%), GPx activity (6.9-fold) and the mRNA expression of antioxidant enzymes Gpx1 (3.9-fold) and Gclc (2.3-fold). Of note, the cytoprotective effect of (PhSe)2 was significantly decreased when cells were treated with mercaptosuccinic acid, an inhibitor of GPx, indicating the involvement of GPx modulation in the observed protective effect. In summary, the present findings bring out a new action mechanism concerning the antioxidant properties of (PhSe)2. The observed upregulation of the glutathione-dependent antioxidant system represents a future pharmacologic possibility that goes beyond the well-known thiol-peroxidase activity of this compound.


Asunto(s)
Derivados del Benceno/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Compuestos de Organoselenio/farmacología , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Glutatión/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Ratones , Modelos Biológicos , Oxidantes/biosíntesis , Oxidación-Reducción/efectos de los fármacos
8.
Free Radic Res ; 51(7-8): 657-668, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28840761

RESUMEN

Organoselenium compounds, such as diphenyl diselenide (PhSe)2 and phenylselenium zinc chloride (PhSeZnCl), show protective activities related to their thiol peroxidase activity. However, depending on experimental conditions, organoselenium compounds can cause toxicity by oxidising thiol groups of proteins and induce the production of reactive oxygen species (ROS). Here, we analysed the toxicity of (PhSe)2 and PhSeZnCl in yeast Saccharomyces cerevisiae. Cell growth of S. cerevisiae after 1, 2, 3, 4, 6, and 16 h of treatment with 2, 4, 6, and 10 µM of (PhSe)2 was evaluated. For comparative purpose, PhSeZnCl was analysed only at 16 h of incubation at equivalent concentrations of selenium (i.e. 4, 8, 12, and 20 µM). ROS production (DCFH-DA), size, granularity, and cell membrane permeability (propidium iodide) were determined by flow cytometry. (PhSe)2 inhibited cell growth at 2 h (10 µM) of incubation, followed by increase in cell size. The increase of cell membrane permeability and granularity (10 µM) was observed after 3 h of incubation, however, ROS production occurs only at 16 h of incubation (10 µM) with (PhSe)2, indicating that ROS overproduction is a more likely consequence of (PhSe)2 toxicity and not its determinant. All tested parameters showed that only concentration of 20 µM induced toxicity in samples incubated with PhSeZnCl. In summary, the results suggest that (PhSe)2 toxicity in S. cerevisiae is time and concentration dependent, presenting more toxicity when compared with PhSeZnCl.


Asunto(s)
Derivados del Benceno/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Compuestos de Organoselenio/farmacología , Especies Reactivas de Oxígeno/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Cloruros/farmacología , Oxidación-Reducción , Compuestos de Selenio/farmacología , Compuestos de Sulfhidrilo/farmacología , Compuestos de Zinc/farmacología
9.
Mater Sci Eng C Mater Biol Appl ; 79: 748-755, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28629076

RESUMEN

The present study investigated stress oxidative parameters and activities of enzymes of the energy metabolism in various brain structures. Rats were subjected to acute and long-term administration of gold nanoparticles (GNPs) with mean diameters of 10nm and 30nm. Adult (60days old) male Wistar rats received a single intraperitoneal injection (acute administration; 70µg·kg-1) or repeated injections once daily for 28days (long-term administration; 70µg·kg-1) of saline solution or GNPs (10nm or 30nm). Twenty-four hours after administration of the final dose, the animals were killed and the cerebral structures were isolated for enzyme analysis. In this study, we observed that the thiobarbituric acid-reactive species and carbonyl protein levels were decreased after acute administration of GNPs, whereas the superoxide dismutase activity was increased after acute and long-term of GNPs. The catalase activity was affected by the administration of GNPs. Furthermore, we have not found change in the citrate synthase activity. The succinate dehydrogenase, malate dehydrogenase, complexes I, II, II-III and IV, and creatine kinase activities were altered. These results indicate that inhibition energy metabolism can be caused by oxidative stress.


Asunto(s)
Nanopartículas del Metal , Animales , Encéfalo , Metabolismo Energético , Oro , Masculino , Estrés Oxidativo , Ratas , Ratas Wistar
10.
Intensive Care Med Exp ; 5(1): 15, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28303482

RESUMEN

BACKGROUND: Extracellular superoxide dismutase (ECSOD) protects nitric oxide (NO) bioavailability by decreasing superoxide levels and preventing peroxynitrite generation, which is important in maintaining renal blood flow and in preventing acute kidney injury. However, the profile of ECSOD expression after sepsis is not fully understood. Therefore, we intended to evaluate the content and gene expression of superoxide dismutase (SOD) isoforms in the renal artery and their relation to renal blood flow. METHODS: Sepsis was induced in Wistar rats by caecal ligation and perforation. Several times after sepsis induction, renal blood flow (12, 24 and 48 h); the renal arterial content of SOD isoforms, nitrotyrosine, endothelial and inducible nitric oxide synthase (e-NOS and i-NOS), and phosphorylated vasodilator-stimulated phosphoprotein (pVASP); and SOD activity (3, 6 and 12 h) were measured. The influence of a SOD inhibitor was also evaluated. RESULTS: An increase in ECSOD content was associated with decreased 3-nitrotyrosine levels. These events were associated with an increase in pVASP content and maintenance of renal blood flow. Moreover, previous treatment with a SOD inhibitor increased nitrotyrosine content and reduced renal blood flow. CONCLUSIONS: ECSOD appears to have a major role in decreasing peroxynitrite formation in the renal artery during the early stages of sepsis development, and its application can be important in renal blood flow control and maintenance during septic insult.

11.
Int J Dev Neurosci ; 48: 24-30, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26608891

RESUMEN

Acute leucine intoxication and neurologic deterioration can develop rapidly at any age as a result of net protein degradation precipitated by infection or psychological stress in patients with maple syrup urine disease (MSUD). Here, we investigated the effects of acute and chronic Hyper-BCAA (H-BCAA) administration on pro- and anti-inflammatory cytokines in the brains of rats. For acute administration, Wistar rats (10 and 30 days) received three injections of BCAA pool (15.8 µL/g at 1-h intervals) or saline, subcutaneously. For chronic administration, Wistar rats (7 days) received of BCAA pool or saline twice a day for 21 days, subcutaneously. Our results showed that acute administration of H-BCAA increased IL-1ß (∼ 78%; p ≤ 0.009) and TNF-α (∼ 155%; p ≤ 0.026) levels in the cerebral cortex but not in the hippocampus of infant rats. Moreover, IL-6 levels were increased in the hippocampus (∼ 135%; p ≤ 0.009) and cerebral cortex (∼ 417%; p ≤ 0.008), whereas IL-10 levels were decreased only in the hippocampus (∼ 42%; p ≤ 0.009). However, repeated administration of H-BCAA decreased IL-1ß (∼ 59%; p ≤ 0.047), IL-6 (∼ 70%; p ≤ 0.009) and IFN-γ (∼ 70%; p ≤ 0.008) levels in the cerebral cortex, whereas the IL-6 (∼ 67%; p ≤ 0.009), IL-10 (∼ 58%; p ≤ 0.01) and IFN-γ (∼ 67%; p ≤ 0.009) levels were decreased in the hippocampus. These findings suggest that a better understanding of the inflammatory response in MSUD patients may be useful to develop therapeutic strategies to modulate the hyperinflammatory/hypoinflammatory axis.


Asunto(s)
Aminoácidos de Cadena Ramificada/administración & dosificación , Encéfalo/efectos de los fármacos , Citocinas/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Esquema de Medicación , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Factores de Tiempo
12.
Mol Neurobiol ; 53(6): 3714-3723, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26133302

RESUMEN

Maple syrup urine disease (MSUD) is a rare metabolic disorder associated with acute and chronic brain dysfunction. This condition has been shown to lead to macroscopic cerebral alterations that are visible on imaging studies. Cerebral oedema is widely considered to be detrimental for MSUD patients; however, the mechanisms involved are still poorly understood. Therefore, we investigated whether acute administration of branched-chain amino acids (BCAA) causes cerebral oedema, modifies the Na(+),K(+)-ATPase activity, affects the permeability of the blood-brain barrier (BBB) and alters the levels of cytokines in the hippocampus and cerebral cortex of 10-day-old rats. Additionally, we investigated the influence of concomitant administration of dexamethasone on the alterations caused by BCAA. Our results showed that the animals submitted to the model of MSUD exhibited an increase in the brain water content, both in the cerebral cortex and in the hippocampus. By investigating the mechanism of cerebral oedema, we discovered an association between H-BCAA and the Na(+),K(+)-ATPase activity and the permeability of the BBB to small molecules. Moreover, the H-BCAA administration increases Il-1ß, IL-6 and TNF-α levels in the hippocampus and cerebral cortex, whereas IL-10 levels were decreased in the hippocampus. Interestingly, we showed that the administration of dexamethasone successfully reduced cerebral oedema, preventing the inhibition of Na(+),K(+)-ATPase activity, BBB breakdown and the increase in the cytokines levels. In conclusion, these findings suggest that dexamethasone can improve the acute cerebral oedema and brain injury associated with high levels of BCAA, either through a direct effect on brain capillary Na(+),K(+)-ATPase or through a generalized effect on the permeability of the BBB to all compounds.


Asunto(s)
Barrera Hematoencefálica/patología , Edema Encefálico/prevención & control , Dexametasona/uso terapéutico , Hipocampo/enzimología , Enfermedad de la Orina de Jarabe de Arce/complicaciones , Enfermedad de la Orina de Jarabe de Arce/tratamiento farmacológico , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Aminoácidos de Cadena Ramificada/administración & dosificación , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/patología , Dexametasona/administración & dosificación , Dexametasona/farmacología , Modelos Animales de Enfermedad , Hipocampo/patología , Masculino , Enfermedad de la Orina de Jarabe de Arce/enzimología , Enfermedad de la Orina de Jarabe de Arce/patología , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo
13.
PLoS One ; 10(5): e0126606, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26010931

RESUMEN

Ethylmalonic acid (EMA) accumulates in tissues and biological fluids of patients affected by short-chain acyl-CoA dehydrogenase deficiency (SCADD) and ethylmalonic encephalopathy, illnesses characterized by neurological and muscular symptoms. Considering that the mechanisms responsible for the brain and skeletal muscle damage in these diseases are poorly known, in the present work we investigated the effects of acute EMA administration on redox status parameters in cerebral cortex and skeletal muscle from 30-day-old rats. Animals received three subcutaneous injections of EMA (6 µmol/g; 90 min interval between injections) and were killed 1 h after the last administration. Control animals received saline in the same volumes. EMA administration significantly increased thiobarbituric acid-reactive substances levels in cerebral cortex and skeletal muscle, indicating increased lipid peroxidation. In addition, carbonyl content was increased in EMA-treated animal skeletal muscle when compared to the saline group. EMA administration also significantly increased 2',7'-dihydrodichlorofluorescein oxidation and superoxide production (reactive species markers), and decreased glutathione peroxidase activity in cerebral cortex, while glutathione levels were decreased only in skeletal muscle. On the other hand, respiratory chain complex I-III activity was altered by acute EMA administration neither in cerebral cortex nor in skeletal muscle. The present results show that acute EMA administration elicits oxidative stress in rat brain and skeletal muscle, suggesting that oxidative damage may be involved in the pathophysiology of the brain and muscle symptoms found in patients affected by SCADD and ethylmalonic encephalopathy.


Asunto(s)
Encéfalo/metabolismo , Malonatos/administración & dosificación , Malonatos/efectos adversos , Músculo Esquelético/metabolismo , Animales , Encéfalo/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Fluoresceínas/metabolismo , Glutatión/metabolismo , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/enzimología , Oxidación-Reducción , Carbonilación Proteica/efectos de los fármacos , Ratas Wistar , Compuestos de Sulfhidrilo/metabolismo , Superóxidos/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
14.
Mol Neurobiol ; 49(2): 1069-76, 2014 04.
Artículo en Inglés | MEDLINE | ID: mdl-24234155

RESUMEN

Sepsis is defined as the host's reaction to infection and characterised by a systemic inflammatory response with important clinical implications. Central nervous system dysfunction secondary to sepsis is associated with local generation of pro- and anti-inflammatory cytokines, impaired cerebral microcirculation, an imbalance of neurotransmitters, apoptosis and cognitive impairment. It's known that the IL-1ß is one of the first cytokines to be altered. Thus, the objective of this study was to evaluate the role of IL-1ß in cognitive parameters in brain tissue through the use of an IL-1ß (IL-1ra) receptor antagonist up to 10 days and to assess blood-brain barrier permeability, cytokine levels, oxidative parameters and energetic metabolism up to 24 h, after sepsis induction. To this aim, we used sham-operated Wistar rats or submitted to the cecal ligation and perforation (CLP) procedure. Immediately after, the animals received one dose of 10 µg of IL-1ra. After 24 h, the rats were killed and were evaluated for biochemical parameters in the pre-frontal cortex, hippocampus and striatum. After 10 days, the animals were submitted to the habituation to the open field and step-down inhibitory avoidance task. We observed that the use of IL-1ra reverted the increase of blood-brain barrier permeability in the pre-frontal cortex, hippocampus and striatum; the increase of IL-1ß, IL1-6 and TNF-α levels in the pre-frontal cortex and striatum; the decrease of complex I activity in the pre-frontal, hippocampus and striatum; the increase of oxidative parameters in pre-frontal cortex, hippocampus and striatum; and cognitive impairment. In conclusion, the results observed in this study reinforce the role of acute brain inflammatory response, in particular, the IL1ß response, in the cognitive impairment associated with sepsis.


Asunto(s)
Trastornos del Conocimiento/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/metabolismo , Sepsis/metabolismo , Animales , Reacción de Prevención/fisiología , Trastornos del Conocimiento/patología , Masculino , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar , Sepsis/psicología
15.
Mol Neurobiol ; 50(2): 358-67, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24390570

RESUMEN

Maple syrup urine disease (MSUD) is an inborn error of metabolism caused by a severe deficiency in the activity of the branched-chain α-keto acid dehydrogenase complex, leading to accumulation of the branched-chain amino acids (BCAA) leucine, isoleucine, and valine. Infections have a significant role in precipitating acute metabolic decompensation in patients with MSUD; however, the mechanisms underlying the neurotoxicity in this disorder are poorly understood. In this study, we subjected rats to the coadministration of lipopolysaccharide (LPS), which is a major component of gram-negative bacteria cell walls, and high concentrations of BCAA (H-BCAA) to determine their effects on the permeability of the blood-brain barrier (BBB) and on the levels of matrix metalloproteinases (MMP-2 and MMP-9). Our results demonstrated that the coadministration of H-BCAA and LPS causes breakdown of the BBB and increases the levels of MMP-2 and MMP-9 in the hippocampus of these rats. On the other hand, examination of the cerebral cortex of the 10- and 30-day-old rats revealed a significant difference in Evan's Blue content after coadministration of H-BCAA and LPS, as MMP-9 levels only increased in the cerebral cortex of the 10-day-old rats. In conclusion, these results suggest that the inflammatory process associated with high levels of BCAA causes BBB breakdown. Thus, we suggest that BBB breakdown is relevant to the perpetuation of brain inflammation and may be related to the brain dysfunction observed in MSUD patients.


Asunto(s)
Aminoácidos de Cadena Ramificada/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Lipopolisacáridos/farmacología , Enfermedad de la Orina de Jarabe de Arce/tratamiento farmacológico , Metaloproteinasas de la Matriz/metabolismo , Aminoácidos de Cadena Ramificada/administración & dosificación , Aminoácidos de Cadena Ramificada/sangre , Animales , Barrera Hematoencefálica/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Hipocampo/efectos de los fármacos , Inflamación/tratamiento farmacológico , Lipopolisacáridos/administración & dosificación , Masculino , Enfermedad de la Orina de Jarabe de Arce/metabolismo , Enfermedad de la Orina de Jarabe de Arce/patología , Ratas Wistar
16.
Pharmacol Biochem Behav ; 103(2): 395-402, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23036485

RESUMEN

Animal models of chronic stress represent valuable tools by which to investigate the behavioral, endocrine and neurobiological changes underlying stress-related psychopathologies, such as major depression, and the efficacy of antidepressant therapies. The present study was aimed at investigating the neurochemical effects of the antidepressant tianeptine in rats exposed to the chronic stress model. To this aim, rats were subjected to 40days of chronic unpredictable stressful stimuli, after which the animals received saline or tianeptine (15mg/kg) once a day for 7days. Additionally, IL-6, IL-1, TNF-α levels and oxidative stress parameters were assessed in the prefrontal cortex (PFC), hippocampus (HPC), amygdala (AMY) and nucleus accumbens (NAc) in all of the experimental groups studied. The results indicated that chronic mild stress and tianeptine did not exercise any effects on cytokines in all of the structures studied; in the PFC and AMY thiobarbituric acid reactive substances (TBARS) levels were decreased in control rats treated with tianeptine in the HPC; superoxide dismutase (SOD) activity was found to have decreased in stressed rats treated with saline in the PFC, HPC, AMY and NAc, and tianeptine reversed this effect; catalase (CAT) activity was found to have decreased in the PFC, HPC and NAc of stressed rats treated with saline, but was shown to have increased in stressed rats treated with tianeptine, and tianeptine also reversed the decreases in CAT activity in stressed rats treated with saline, suggesting that tianeptine exerted antioxidant activity. In conclusion, the present findings open new vistas on the pharmacological activity of tianeptine, in particular, concerning its ability to attenuate oxidative stress.


Asunto(s)
Encéfalo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Psicológico , Tiazepinas/farmacología , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Catalasa/metabolismo , Enfermedad Crónica , Masculino , Estrés Oxidativo , Ratas , Ratas Wistar , Superóxido Dismutasa/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda