Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Acta Neurobiol Exp (Wars) ; 83(2): 216-225, 2023.
Article in English | MEDLINE | ID: mdl-37493537

ABSTRACT

This study investigated the effects of sub­chronic administration of lead (Pb) acetate on thiobarbituric acid reactive substances (TBA­RS), total sulfhydryl content, protein carbonyl content, antioxidant enzymes (superoxide dismutase [SOD], catalase [CAT], glutathione peroxidase [GSH­Px]), acetylcholinesterase (AChE), and Na+K+­ATPase in the cerebral structures of rats. Male Wistar rats aged 60 days were treated with saline (control group) or Pb (treatment group), at various doses, by gavage, once a day for 35 days. The animals were sacrificed twelve hours after the last administration, and the cerebellum, hippocampus and cerebral cortex were removed. The results showed that Pb did not alter the evaluated oxidative stress parameters. Furthermore, Pb (64 and/or 128 mg/kg) altered SOD in the cerebellum, cerebral cortex and hippocampus. Pb (128 mg/kg) altered CAT in the cerebellum and cerebral cortex and GSH­Px in the cerebral cortex. Also, Pb (64 mg/kg and 128 mg/kg) altered GSH­Px in the cerebellum. Moreover, Pb (128 mg/kg) increased AChE in the hippocampus and decreased Na+K+­ATPase in the cerebellum and hippocampus. In conclusion, sub­chronic exposure to Pb (occupational and environmental intoxication) altered antioxidant enzymes, AChE, and Na+K+­ATPase, contributing to cerebral dysfunction.


Subject(s)
Acetylcholinesterase , Antioxidants , Rats , Male , Animals , Antioxidants/metabolism , Acetylcholinesterase/metabolism , Rats, Wistar , Protein Carbonylation , Lead/toxicity , Lead/metabolism , Oxidative Stress , Catalase/metabolism , Cerebral Cortex/metabolism , Superoxide Dismutase/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/pharmacology , Brain/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Thiobarbituric Acid Reactive Substances/pharmacology
2.
Metab Brain Dis ; 35(5): 765-774, 2020 06.
Article in English | MEDLINE | ID: mdl-32189127

ABSTRACT

During chronic inflammatory disease, such asthma, leukocytes can invade the central nervous system (CNS) and together with CNS-resident cells, generate excessive reactive oxygen species (ROS) production as well as disbalance in the antioxidant system, causing oxidative stress, which contributes a large part to neuroinflammation. In this sense, the aim of this study is to investigate the effects of treatment with neostigmine, known for the ability to control lung inflammation, on oxidative stress in the cerebral cortex of asthmatic mice. Female BALB/cJ mice were submitted to asthma model induced by ovalbumin (OVA). Control group received only Dulbecco's phosphate-buffered saline (DPBS). To evaluate neostigmine effects, mice received 80 µg/kg of neostigmine intraperitoneally 30 min after each OVA challenge. Our results revealed for the first time that treatment with neostigmine (an acetylcholinesterase inhibitor that no crosses the BBB) was able to revert ROS production and change anti-oxidant enzyme catalase in the cerebral cortex in asthmatic mice. These results support the communication between the peripheral immune system and the CNS and suggest that acetylcholinesterase inhibitors, such as neostigmine, should be further studied as possible therapeutic strategies for neuroprotection in asthma.


Subject(s)
Asthma/drug therapy , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cholinesterase Inhibitors/pharmacology , Neostigmine/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Asthma/chemically induced , Asthma/pathology , Bronchoalveolar Lavage Fluid , Catalase/metabolism , Cholinesterase Inhibitors/therapeutic use , Female , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Neostigmine/therapeutic use , Neuroprotection , Neuroprotective Agents/therapeutic use , Ovalbumin , Reactive Oxygen Species/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Superoxide Dismutase-1/metabolism
3.
J Cell Physiol ; 232(12): 3552-3564, 2017 12.
Article in English | MEDLINE | ID: mdl-28112391

ABSTRACT

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are syndromes of acute hypoxemic respiratory failure resulting from a variety of direct and indirect injuries to the gas exchange parenchyma of the lungs. During the ALI, we have an increase release of proinflammatory cytokines and high reactive oxygen species (ROS) formation. These factors are responsible for the release and activation of neutrophil-derived proteases and the formation of neutrophil extracellular traps (NETs). The excessive increase in the release of NETs cause damage to lung tissue. Recent studies have studies involving the administration of mesenchymal stem cells (MSCs) for the treatment of experimental ALI has shown promising results. In this way, the objective of our study is to evaluate the ability of MSCs, in a lipopolysaccharide (LPS)-induced ALI model, to reduce inflammation, oxidative damage, and consequently decrease the release of NETs. Mice were submitted lung injury induced by intratracheal instillation of LPS and subsequently treated or not with MSCs. Treatment with MSCs was able to modulate pulmonary inflammation, decrease oxidative damage, and reduce the release of NETs. These benefits from treatment are evident when we observe a significant increase in the survival curve in the treated animals. Our results demonstrate that MSCs treatment is effective for the treatment of ALI. For the first time, it is described that MSCs can reduce the formation of NETs and an experimental model of ALI. This finding is directly related to these cells modulate the inflammatory response and oxidative damage in the course of the pathology.


Subject(s)
Acute Lung Injury/surgery , Extracellular Traps/metabolism , Lung/metabolism , Mesenchymal Stem Cell Transplantation , Pneumonia/surgery , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Cells, Cultured , Chemotaxis , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Lipopolysaccharides , Lung/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/pathology , Oxidative Stress , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology , Time Factors
4.
J Biochem Mol Toxicol ; 31(1): 1-7, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27489181

ABSTRACT

We investigated the in vitro and in vivo effects of arginine (Arg) on thiobarbituric acid-reactive substances (TBA-RS) and on the activities of catalase (CAT), glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) in renal tissues of rats. We also studied the influence of antioxidants (α-tocopherol plus ascorbic acid) and nitric oxide synthase inhibitor NG -nitro-l-arginine methyl ester (l-NAME) on the effects elicited by Arg. Results showed that Arg in vitro (1.5 mM) decreased SOD activity and increased the levels of TBA-RS in the renal medulla. Acute administration of Arg [0.8 g/kg, intraperitoneal injection] decreased CAT activity, increased SOD activity and TBA-RS levels in the renal medulla, and decreased CAT activity in the renal cortex of rats. Most results were prevented by antioxidants and/or l-NAME. Data indicate that Arg causes an oxidative imbalance in the renal tissues studied; however, in the presence of antioxidants and l-NAME, some of these alterations in oxidative stress were prevented.


Subject(s)
Antioxidants/pharmacology , Hyperargininemia/prevention & control , Kidney/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Oxidative Stress/drug effects , alpha-Tocopherol/pharmacology , Animals , Catalase/metabolism , Glutathione Peroxidase/metabolism , Hyperargininemia/chemically induced , Hyperargininemia/metabolism , Male , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
5.
Mol Cell Biochem ; 413(1-2): 47-55, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26738487

ABSTRACT

The inflammatory cells infiltrating the airways produce several mediators, such as reactive oxygen species (ROS). ROS and the oxidant-antioxidant imbalance might play an important role in the modulation of airways inflammation. In order to avoid the undesirable effects of ROS, various endogenous antioxidant strategies have evolved, incorporating both enzymatic and non-enzymatic mechanisms. Recombinant human deoxyribonuclease (rhDNase) in clinical studies demonstrated a reduction in sputum viscosity, cleaving extracellular DNA in the airways, and facilitating mucus clearance, but an antioxidant effect was not studied so far. Therefore, we evaluated whether the administration of rhDNase improves oxidative stress in a murine model of asthma. Mice were sensitized by two subcutaneous injections of ovalbumin (OVA), on days 0 and 7, followed by three lung challenges with OVA on days 14, 15, and 16. On days 15 and 16, after 2 h of the challenge with OVA, mice received 1 mg/mL of rhDNase in the lungs. Bronchoalveolar lavage fluid and lung tissue were obtained on day 17, for inflammatory and oxidative stress analysis. We showed that rhDNase did not alter the population of inflammatory cells, such as eosinophil cells, in OVA-treated rhDNase group but significantly improved oxidative stress in lung tissue, by decreasing oxygen reactive species and increasing superoxide dismutase/catalase ratio, glutathione peroxidase activity, and thiol content. Our data provide the first evidence that rhDNase decreases some measures of oxidative stress and antioxidant status in a murine model of asthma, with a potential antioxidant effect to be further studied in human asthma.


Subject(s)
Asthma/immunology , Deoxyribonucleases/administration & dosage , Eosinophils/metabolism , Lung/immunology , Oxidative Stress/drug effects , Animals , Asthma/chemically induced , Asthma/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Deoxyribonucleases/genetics , Deoxyribonucleases/metabolism , Disease Models, Animal , Female , Humans , Mice , Ovalbumin/adverse effects , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
6.
Purinergic Signal ; 11(4): 463-70, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26265456

ABSTRACT

Elevated plasma levels of homocysteine (Hcy) are associated with the development of coronary artery disease (CAD), peripheral vascular disease, and atherosclerosis. Hyperhomocysteinemia is likely related to the enhanced production of pro-inflammatory cytokines including IL-1ß. However, the mechanisms underlying the effects of Hcy in immune cells are not completely understood. Recent studies have established a link between macrophage accumulation, cytokine IL-1ß, and the advance of vascular diseases. The purpose of the present study is to investigate the effects of Hcy on IL-1ß secretion by murine macrophages. Hcy (100 µM) increases IL-1ß synthesis via enhancement of P2X7 expression and NF-ĸB and ERK activation in murine macrophages. In addition, the antioxidant agent N-acetylcysteine (NAC) reduces NF-κB activation, ERK phosphorylation, and IL-1ß production in Hcy-exposed macrophages, indicating the importance of ROS in this pro-inflammatory process. In summary, our results show that Hcy may be involved in the synthesis and secretion of IL-1ß via NF-ĸB, ERK, and P2X7 stimulation in murine macrophages.


Subject(s)
Homocysteine/toxicity , Hyperhomocysteinemia/metabolism , Interleukin-1beta/biosynthesis , MAP Kinase Signaling System/drug effects , Macrophages/metabolism , NF-kappa B/metabolism , Receptors, Purinergic P2X7/metabolism , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/genetics
7.
Gene ; 531(2): 191-8, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24035933

ABSTRACT

Sulfite oxidase (SO) deficiency is biochemically characterized by tissue accumulation and high urinary excretion of sulfite, thiosulfate and S-sulfocysteine. Affected patients present severe neurological symptoms and cortical atrophy, whose pathophysiology is still poorly established. Therefore, in the present work we investigated the in vitro effects of sulfite and thiosulfate on important parameters of energy metabolism in the brain of young rats. We verified that sulfite moderately inhibited the activity of complex IV, whereas thiosulfate did not alter any of the activities of the respiratory chain complexes. It was also found that sulfite and thiosulfate markedly reduced the activity of total creatine kinase (CK) and its mitochondrial and cytosolic isoforms, suggesting that these metabolites impair brain cellular energy buffering and transfer. In contrast, the activity of synaptic Na(+),K(+)-ATPase was not altered by sulfite or thiosulfate. We also observed that the inhibitory effect of sulfite and thiosulfate on CK activity was prevented by melatonin, reduced glutathione and the combination of both antioxidants, as well as by the nitric oxide synthase N(ω)-nitro-l-arginine methyl ester, indicating the involvement of reactive oxygen and nitrogen species in these effects. Sulfite and thiosulfate also increased 2',7'-dichlorofluorescin oxidation and hydrogen peroxide production and decreased the activity of the redox sensor aconitase enzyme, reinforcing a role for oxidative damage in the effects elicited by these metabolites. It may be presumed that the disturbance of cellular energy and redox homeostasis provoked by sulfite and thiosulfate contributes to the neurological symptoms and abnormalities found in patients affected by SO deficiency.


Subject(s)
Amino Acid Metabolism, Inborn Errors/complications , Brain Diseases, Metabolic/etiology , Brain/drug effects , Energy Metabolism/drug effects , Homeostasis/drug effects , Sulfite Oxidase/deficiency , Sulfites/pharmacology , Thiosulfates/pharmacology , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Brain/metabolism , Brain/pathology , Brain/physiology , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/metabolism , Electron Transport/drug effects , Electron Transport/genetics , Electron Transport/physiology , Energy Metabolism/physiology , Male , Oxidation-Reduction/drug effects , Rats , Rats, Wistar , Sulfite Oxidase/genetics , Sulfite Oxidase/metabolism , Sulfites/metabolism , Thiosulfates/metabolism
8.
Neurochem Res ; 38(11): 2342-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24013887

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder whose pathogenesis involves production and aggregation of amyloid-ß peptide (Aß). Aß-induced toxicity is believed to involve alterations on as Na(+),K(+)-ATPase and acetylcholinesterase (AChE) activities, prior to neuronal death. Drugs able to prevent or to reverse these biochemical changes promote neuroprotection. GM1 is a ganglioside proposed to have neuroprotective roles in AD models, through mechanisms not yet fully understood. Therefore, this study aimed to investigate the effect of Aß1-42 infusion and GM1 treatment on recognition memory and on Na(+),K(+)-ATPase and AChE activities, as well as, on antioxidant defense in the brain cortex and the hippocampus. For these purposes, Wistar rats received i.c.v. infusion of fibrilar Aß1-42 (2 nmol) and/or GM1 (0.30 mg/kg). Behavioral and biochemical analyses were conducted 1 month after the infusion procedures. Our results showed that GM1 treatment prevented Aß-induced cognitive deficit, corroborating its neuroprotective function. Aß impaired Na(+),K(+)-ATPase and increase AChE activities in hippocampus and cortex, respectively. GM1, in turn, has partially prevented Aß-induced alteration on Na(+),K(+)-ATPase, though with no impact on AChE activity. Aß caused a decrease in antioxidant defense, specifically in hippocampus, an effect that was prevented by GM1 treatment. GM1, both in cortex and hippocampus, was able to increase antioxidant scavenge capacity. Our results suggest that Aß-triggered cognitive deficit involves region-specific alterations on Na(+),K(+)-ATPase and AChE activities, and that GM1 neuroprotection involves modulation of Na(+),K(+)-ATPase, maybe by its antioxidant properties. Although extrapolation from animal findings is difficult, it is conceivable that GM1 could play an important role in AD treatment.


Subject(s)
Acetylcholinesterase/metabolism , Amyloid beta-Peptides/pharmacology , G(M1) Ganglioside/pharmacology , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Acetylcholinesterase/drug effects , Animals , Injections, Intraventricular , Male , Memory/drug effects , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/drug effects
9.
Toxicol In Vitro ; 27(8): 2273-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24055814

ABSTRACT

Increased levels of plasma homocysteine (hyperhomocysteinemia-HHcy) are associated to the development of coronary artery disease (CAD), peripheral vascular disease and thrombosis. In addition, recent studies have shown that inflammation, probably mediated by macrophages, mediates the pathogenesis associated to high levels of homocysteine (Hcy). In the present study, we evaluated the Hcy effects in the ATP hydrolysis and its breakdown products in murine macrophages. The results showed that micromolar concentrations of Hcy increased the ATP, ADP and AMP hydrolysis. Additionally, our results show decreased inosine levels in the extracellular milieu of Hcy-exposed macrophages. The increasing in ATP, ADP and AMP hydrolysis are not explained by increased transcription or protein expression of NTPDases and ecto-5'-nucleotidase (ecto-5'-NT/CD73) enzymes. Moreover, the formation of reactive oxygen species did not interfere in the Hcy effects, which suggest that Hcy or Hcy metabolites act directly on the modulation of NTPDases and ecto-5'-NT/CD73 activities. In conclusion, Hcy induces the rapid breakdown of ATP, ADP and AMP to adenosine (ADO), which is classically known as an anti-inflammatory response in immune cells. However, by the action of these enzymes, the extracellular adenosine generated during Hcy treatment probably is uptaken into the cells, as evidenced by the decreased in inosine formation, and thus collaborating to the inflammatory complications associates to HHcy.


Subject(s)
Adenosine Triphosphate/metabolism , Homocysteine/pharmacology , Macrophages, Peritoneal/drug effects , Adenosine Triphosphatases/genetics , Animals , Apyrase/genetics , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Mutant Strains , Pyrophosphatases/genetics , RNA, Messenger/metabolism , Receptors, Purinergic P1/genetics , Receptors, Purinergic P2/genetics
10.
Mol Cell Biochem ; 384(1-2): 21-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23963990

ABSTRACT

It has been shown that elevation of plasma methionine (Met) and its metabolites may occur in several genetic abnormalities. In this study we investigated the in vitro and in vivo effects of the Met and methionine sulfoxide (MetO) on oxidative stress parameters in the liver of rats. For in vitro studies, liver homogenates were incubated with Met, MetO, and Mix (Met + MetO). For in vivo studies, the animals were divided into groups: saline, Met 0.4 g/kg, MetO 0.1 g/kg, and Met 0.4 g/kg + MetO 0.1 g/kg. The animals were euthanized 1 and 3 h after injection. In vitro results showed that Met 1 and 2 mM and Mix increased catalase (CAT) activity. Superoxide dismutase (SOD) was enhanced by Met 1 and 2 mM, MetO 0.5 mM, and Mix. Dichlorofluorescein oxidation was increased by Met 1 mM and Mix. In vivo results showed that Met, MetO, and Mix decreased TBARS levels at 1 h. Total thiol content decreased 1 h after and increased 3 h after MetO and Met plus MetO administrations. Carbonyl content was enhanced by Met and was reduced by MetO 1 h after administration. Met, MetO and Met plus MetO decreased CAT activity 1 and 3 h after administration. Furthermore, only MetO increased SOD activity. In addition, Met, MetO, and Mix decreased dichlorofluorescein oxidation at 1 and 3 h. Our data indicate that Met/MetO in vivo and in vitro modify liver homeostasis by altering the redox cellular state. However, the hepatic changes caused by these compounds suggest a short-time adaptation of this tissue.


Subject(s)
Catalase/metabolism , Liver/metabolism , Methionine/analogs & derivatives , Methionine/metabolism , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Fluoresceins/metabolism , Glycine N-Methyltransferase/deficiency , Glycine N-Methyltransferase/metabolism , Liver/pathology , Male , Oxidative Stress , Rats , Rats, Wistar , Reactive Oxygen Species/pharmacology , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
11.
Mol Neurobiol ; 46(2): 467-74, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22810802

ABSTRACT

In the present study, we investigated whether sepsis induced by cecal ligation and puncture (CLP) modifies Na(+), K(+)-ATPase activity, mRNA expression, and cerebral edema in hippocampus and cerebral cortex of rats and if antioxidant (ATX) treatment prevented the alterations induced by sepsis. Rats were subjected to CLP and were divided into three groups: sham; CLP-rats were subjected to CLP without any further treatment; and ATX-CLP plus administration of N-acetylcysteine plus deferoxamine. Several times (6, 12, and 24) after CLP or sham operation, the rats were killed and hippocampus and cerebral cortex were isolated. Na(+), K(+)-ATPase activity was inhibited in the hippocampus 24 h after sepsis, and ATX treatment was not able to prevent this inhibition. The Na(+), K(+)-ATPase activity also was inhibited in cerebral cortex 6, 12, and 24 h after sepsis. No differences on Na(+), K(+)-ATPase catalytic subunit mRNA levels were found in the hippocampus and cerebral cortex after sepsis. ATX treatment prevents Na(+), K(+)-ATPase inhibition only in the cerebral cortex. Na(+), K(+)-ATPase inhibition was not associated to increase brain water content. In conclusion, the present study demonstrated that sepsis induced by CLP inhibits Na(+), K(+)-ATPase activity in a mechanism dependent on oxidative stress, but this is not associated to increase brain water content.


Subject(s)
Antioxidants/pharmacology , Cerebral Cortex/enzymology , Hippocampus/enzymology , Sepsis/enzymology , Sepsis/pathology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Catalytic Domain , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Disease Models, Animal , Gene Expression Regulation, Enzymologic/drug effects , Hippocampus/drug effects , Hippocampus/pathology , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sepsis/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Water/metabolism
12.
PLoS One ; 7(2): e31205, 2012.
Article in English | MEDLINE | ID: mdl-22348056

ABSTRACT

Macrophages are key elements in the inflammatory process, whereas depending on the micro-environmental stimulation they exhibit a pro-inflammatory (classical/M1) or an anti-inflammatory/reparatory (alternative/M2) phenotype. Extracellular ATP can act as a danger signal whereas adenosine generally serves as a negative feedback mechanism to limit inflammation. The local increase in nucleotides communication is controlled by ectonucleotidases, such as members of the ectonucleoside triphosphate diphosphohydrolase (E-NTPDase) family and ecto-5'-nucleotidase/CD73 (ecto-5'-NT). In the present work we evaluated the presence of these enzymes in resident mice M1 (macrophages stimulated with LPS), and M2 (macrophages stimulated with IL-4) macrophages. Macrophages were collected by a lavage of the mice (6-8 weeks) peritoneal cavity and treated for 24 h with IL-4 (10 ng/mL) or LPS (10 ng/mL). Nitrite concentrations were measured using the Greiss reaction. Supernatants were harvested to determine cytokines and the ATPase, ADPase and AMPase activities were determined by the malachite green method and HPLC analysis. The expression of selected surface proteins was evaluated by flow cytometry. The results reveal that M1 macrophages presented a decreased ATP and AMP hydrolysis in agreement with a decrease in NTPDase1, -3 and ecto-5'-nucleotidase expression compared to M2. In contrast, M2 macrophages showed a higher ATP and AMP hydrolysis and increased NTPDase1, -3 and ecto-5'-nucleotidase expression compared to M1 macrophages. Therefore, macrophages of the M1 phenotype lead to an accumulation of ATP while macrophages of the M2 phenotype may rapidly convert ATP to adenosine. The results also showed that P1 and P2 purinoreceptors present the same mRNA profile in both phenotypes. In addition, M2 macrophages, which have a higher ATPase activity, were less sensitive to cell death. In conclusion, these changes in ectoenzyme activities might allow macrophages to adjust the outcome of the extracellular purinergic cascade in order to fine-tune their functions during the inflammatory set.


Subject(s)
5'-Nucleotidase/analysis , Gene Expression Profiling , Macrophage Activation/genetics , Pyrophosphatases/analysis , 5'-Nucleotidase/genetics , Adenosine/biosynthesis , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Animals , Mice , Pyrophosphatases/genetics , RNA, Messenger/analysis , Receptors, Purinergic P1/genetics , Receptors, Purinergic P2/genetics
13.
Stem Cells Dev ; 20(7): 1171-81, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20955077

ABSTRACT

Cell therapy using bone marrow-derived mesenchymal stem cells (MSCs) seems to be a new alternative for the treatment of neurodegenerative diseases. Despite several promising results with their use, possible side effects are still unknown. In a previous work, we have shown that MSC-conditioned medium is toxic to hippocampal slice cultures and aggravates cell death induced by oxygen and glucose deprivation. In this work, we investigated whether the inflammatory response and/or reactive species formation could be involved in that toxicity. Rat organotypic hippocampal cultures were exposed for 24 h to conditioned medium from MSCs isolated from rat bone marrow. A marked glial activation was observed after exposure of cultures to MSC-conditioned medium, as evidenced by glial fibrillary acid protein (GFAP) and isolectin B(4) increase. Tumor necrosis factor-α and interleukin-6 levels were increased in the culture medium, and 2,7-dihydrodichlorofluorescein diacetate oxidation (indicating reactive species generation) and inducible nitric oxide synthase (iNOS) immunocontent were also higher after exposure of cultures to MSC-conditioned medium. Antioxidants (ascorbic acid and TROLOX(®)), N(ω)-nitro-l-arginine methyl ester hydrochloride, and anti-inflammatory drugs (indomethacin and dexamethasone) reduced cell death in hippocampal organotypic cultures after their exposure to MSC-conditioned medium. The results obtained here suggest that MSC-secreted factors trigger reactive species generation and neuroinflammation in organotypic cultures of hippocampus, introducing a note of caution in the use of these cells for neurological application.


Subject(s)
Culture Media, Conditioned/pharmacology , Hippocampus/drug effects , Mesenchymal Stem Cells/metabolism , Neurogenic Inflammation/metabolism , Reactive Oxygen Species/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Blotting, Western , Bone Marrow Cells/metabolism , Cell Death , Cells, Cultured , Glycoproteins/metabolism , Hippocampus/cytology , In Vitro Techniques , Interleukins/analysis , Lectins/metabolism , Male , Neuroglia/drug effects , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/analysis , Versicans
14.
Metab Brain Dis ; 25(2): 145-54, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20431931

ABSTRACT

5-Oxoproline accumulates in glutathione synthetase deficiency, an autossomic recessive inherited disorder clinically characterized by hemolytic anemia, metabolic acidosis, and severe neurological symptoms whose mechanisms are poorly known. In the present study we investigated the effects of acute subcutaneous administration of 5-oxoproline to verify whether oxidative stress is elicited by this metabolite in vivo in cerebral cortex and cerebellum of 14-day-old rats. Our results showed that the acute administration of 5-oxoproline is able to promote both lipid and protein oxidation, to impair brain antioxidant defenses, to alter SH/SS ratio and to enhance hydrogen peroxide content, thus promoting oxidative stress in vivo, a mechanism that may be involved in the neuropathology of gluthatione synthetase deficiency.


Subject(s)
Antioxidants/metabolism , Brain Diseases, Metabolic, Inborn/chemically induced , Cerebellum/drug effects , Cerebrum/drug effects , Lipid Peroxidation/drug effects , Nerve Tissue Proteins/metabolism , Oxidative Stress/drug effects , Pyrrolidonecarboxylic Acid/toxicity , Age Factors , Animals , Antioxidants/physiology , Brain Diseases, Metabolic, Inborn/metabolism , Cerebellum/metabolism , Cerebrum/metabolism , Disease Models, Animal , Glutathione Synthase/deficiency , Lipid Peroxidation/physiology , Nerve Tissue Proteins/physiology , Oxidative Stress/physiology , Pyrrolidonecarboxylic Acid/metabolism , Rats , Rats, Wistar
15.
Cell Mol Neurobiol ; 30(4): 557-68, 2010 May.
Article in English | MEDLINE | ID: mdl-19937110

ABSTRACT

In this study, we investigated the actions of high homocysteine (Hcy) levels (100 and 500 microM) on the cytoskeleton of C6 glioma cells. Results showed that the predominant cytoskeletal response was massive formation of actin-containing filopodia at the cell surface that could be related with Cdc42 activation and increased vinculin immunocontent. In cells treated with 100 microM Hcy, folic acid, trolox, and ascorbic acid, totally prevented filopodia formation, while filopodia induced by 500 microM Hcy were prevented by ascorbic acid and attenuated by folic acid and trolox. Moreover, competitive NMDA ionotropic antagonist DL-AP5 totally prevented the formation of filopodia in both 100 and 500 microM Hcy treated cells, while the metabotropic non-selective group I/II antagonist MCPG prevented the effect of 100 microM Hcy but only slightly attenuated the effect induced by of 500 microM Hcy on actin cytoskeleton. The competitive non-NMDA ionotropic antagonist CNQX was not able to prevent the effects of Hcy on the reorganization of actin cytoskeleton in the two concentrations used. Also, Hcy-induced hypophosphorylation of vimentin and glial fibrillary acidic protein (GFAP) and this effect was prevented by DL-AP5, MCPG, and CNQX. In conclusion, our results show that Hcy target the cytoskeleton of C6 cells probably by excitoxicity and/or oxidative stress mechanisms. Therefore, we could propose that the dynamic restructuring of the actin cytoskeleton of glial cells might contribute to the response to the injury provoked by elevated Hcy levels in brain.


Subject(s)
Actins/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Homocysteine/pharmacology , Intermediate Filaments/metabolism , Neuroglia/cytology , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Cell Line , Chromans/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Folic Acid/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Phosphorylation , Rats , Vimentin/metabolism , Vitamin B Complex/pharmacology
16.
Int J Dev Neurosci ; 26(3-4): 293-9, 2008.
Article in English | MEDLINE | ID: mdl-18329219

ABSTRACT

3-Hydroxyisobutyric aciduria is an inherited metabolic disease caused by 3-hydroxyisobutyryl-CoA dehydrogenase deficiency. Tissue accumulation and high urinary excretion of 3-hydroxyisobutyric acid is the biochemical hallmark of this disorder. Clinical phenotype is heterogeneous and generally includes dysmorphic features, delayed motor development, profound mental impairment, and acute encephalopathy. Lactic acidemia is also found in the affected patients, indicating that mitochondrial dysfunction may be involved in the pathophysiology of this disorder. Therefore, the aim of the present work was to investigate the in vitro effect of 3-hydroxyisobutyric acid (0.1, 0.5 and 1mM) on essential enzymes of energy metabolism, namely the activities of the respiratory chain complexes I-V, total, cytosolic and mitochondrial creatine kinase and Na(+), K(+)-ATPase in cerebral cortex homogenates of 30-day-old rats. We also measured the rate of oxygen consumption in brain mitochondrial preparations in the presence of 3-hydroxyisobutyric acid. 3-Hydroxyisobutyric acid significantly reduced complex I-III (20%), without affecting the other activities of the electron transport chain. Furthermore, 3-hydroxyisobutyric acid did not change state III, state IV and the respiratory control ratio in the presence of glutamate/malate or succinate, suggesting that its effect on cellular respiration was weak. On the other hand, the activities of total and mitochondrial creatine kinase, but not cytosolic creatine kinase, were inhibited (30%) by 3-hydroxyisobutyric acid. We also observed that 3-hydroxyisobutyric acid-induced inhibition of mitochondrial creatine kinase activity was fully prevented by pre-incubation of the homogenates with reduced glutathione, alpha-tocopherol or the combination of superoxide dismutase plus catalase, suggesting that this inhibition was mediated by oxidation of essential thiol groups of the enzyme probably by superoxide, hydrogen peroxide and/or peroxyl radicals. It was also demonstrated that Na(+), K(+)-ATPase activity from synaptic plasma membranes was markedly suppressed (37%) by 3-hydroxyisobutyric acid and that this effect was prevented by alpha-tocopherol co-incubation implying that peroxyl radicals were probably involved in this action. Considering the importance of the affected enzyme activities for brain metabolism homeostasis and neurotransmision, it is suggested that increased tissue levels of 3-hydroxyisobutyric acid may contribute to the neurodegeneration of patients affected by 3-hydroxyisobutyric aciduria and possibly explain previous reports describing elevated production and excretion of lactate.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Brain Diseases, Metabolic, Inborn/enzymology , Cerebral Cortex/enzymology , Energy Metabolism/physiology , 3-Hydroxybutyric Acid/pharmacology , Aging/metabolism , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Brain Chemistry/drug effects , Brain Diseases, Metabolic, Inborn/genetics , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Respiration/drug effects , Cell Respiration/physiology , Cerebral Cortex/drug effects , Creatine Kinase/drug effects , Creatine Kinase/metabolism , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Energy Metabolism/drug effects , Mitochondria/enzymology , Mitochondria/ultrastructure , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Subcellular Fractions , Synaptic Membranes/drug effects , Synaptic Membranes/enzymology
17.
Neurochem Int ; 52(6): 1276-83, 2008 May.
Article in English | MEDLINE | ID: mdl-18295933

ABSTRACT

We previously demonstrated that intrastriatal injection of hypoxanthine, the major metabolite accumulating in Lesch-Nyhan disease, inhibited Na+,K+-ATPase activity and induced oxidative stress in rat striatum. In the present study, we evaluated the action of vitamins E and C on the biochemical alteration induced by hypoxanthine administration on Na+,K+-ATPase, TBARS, TRAP, as well as on superoxide dismutase (SOD), catalase (CAT) and glutathione-peroxidase (GPx) activities in striatum of adult rats. Animals received pretreatment with vitamins E and C or saline during 7 days. Twelve hours after the last injection of vitamins or saline, animals were divided into two groups: (1) vehicle-injected group and (2) hypoxanthine-injected group. For all parameters investigated in this research, animals were sacrificed 30 min after drug infusion. Results showed that pretreatment with vitamins E and C prevented hypoxanthine-mediated effects on Na+,K+-ATPase, TBARS and antioxidant enzymes (SOD, CAT and GPx) activities; however the reduction on TRAP was not prevented by these vitamins. Although extrapolation of findings from animal experiments to humans is difficult, it is conceivable that these vitamins might serve as an adjuvant therapy in order to avoid progression of striatal damage in patients affected by Lesch-Nyhan disease.


Subject(s)
Ascorbic Acid/pharmacology , Corpus Striatum/drug effects , Hypoxanthine/antagonists & inhibitors , Lesch-Nyhan Syndrome/drug therapy , Oxidative Stress/drug effects , Vitamin E/pharmacology , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Ascorbic Acid/therapeutic use , Catalase/drug effects , Catalase/metabolism , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Progression , Free Radicals/metabolism , Hypoxanthine/metabolism , Hypoxanthine/toxicity , Lesch-Nyhan Syndrome/metabolism , Lesch-Nyhan Syndrome/physiopathology , Oxidative Stress/physiology , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Thiobarbituric Acid Reactive Substances/metabolism , Treatment Outcome , Vitamin E/metabolism , Vitamin E/therapeutic use
18.
Neurochem Res ; 33(6): 1129-37, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18256932

ABSTRACT

Guanidinoacetate methyltransferase (GAMT) deficiency is a disorder of creatine metabolism characterized by low plasma creatine concentrations in combination with elevated guanidinoacetate (GAA) concentrations. The aim of this work was to investigate the in vitro effect of guanidinoacetate in NTPDase, 5'-nucleotidase and acetylcholinesterase activities in the synaptosomes, platelets and blood of rats. The results showed that in synaptosomes the NTPDase and 5'-nucleotidase activities were inhibited significantly in the presence of GAA at concentrations of 50, 100, 150 and 200 microM (P < 0.05). However, in platelets GAA at the same concentrations caused a significant increase in the activities of these two enzymes (P < 0.05). In relation to the acetylcholinesterase activity, GAA caused a significant inhibition in the activity of this enzyme in blood at concentrations of 150 and 200 microM (P < 0.05), but did not alter the acetylcholinesterase activity in synaptosomes from the cerebral cortex. Our results suggest that alterations caused by GAA in the activities of these enzymes may contribute to the understanding of the neurological dysfunction of GAMT-deficient patients.


Subject(s)
5'-Nucleotidase/metabolism , Acetylcholinesterase/metabolism , Adenine Nucleotides/metabolism , Antigens, CD/metabolism , Apyrase/metabolism , Glycine/analogs & derivatives , Animals , Brain/metabolism , Glycine/metabolism , Guanidinoacetate N-Methyltransferase/metabolism , Hydrolysis , Isoenzymes/metabolism , Male , Rats , Rats, Wistar , Synaptosomes/metabolism
19.
Neurochem Res ; 33(5): 737-44, 2008 May.
Article in English | MEDLINE | ID: mdl-17940891

ABSTRACT

Cysteamine is a cystine-depleting drug used in the treatment of cystinosis, a metabolic disorder caused by deficiency of the lysosomal cystine carrier. As a result, cystine accumulates within lysosomes in many tissues and organs, including the nervous system. Studies with cystine dimethyl ester loaded cells suggest that cystine might induce apoptosis through oxidative stress. Our objective was to investigate the effects of co-administration of cysteamine with the oxidant cystine dimethyl ester on several parameters of oxidative stress in the brain cortex of rats. Animals were injected with 1.6 micromol/g cystine dimethyl ester and/or 0.26 micromol/g body weight cysteamine. Cystine dimethyl ester induced lipoperoxidation, protein carbonylation, and stimulated superoxide dismutase, glutathione peroxidase and catalase activities, probably through the formation of free radicals. Cysteamine prevented those effects, possibly increasing cellular thiol pool and acting as a scavenger of free radicals. These results suggest that the antioxidant effect of cysteamine may be important in the treatment of cystinosis.


Subject(s)
Antioxidants/pharmacology , Cerebral Cortex/drug effects , Cysteamine/pharmacology , Animals , Catalase/metabolism , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Glutathione Peroxidase/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
20.
Curr Neurovasc Res ; 4(3): 184-93, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17691972

ABSTRACT

Thyroid hormones (THs), including triiodothyronine (T3) and tetraiodothyronine (T4), are recognized as key metabolic hormones of the body. THs are essential for normal maturation and function of the mammalian central nervous system (CNS) and its deficiency, during a critical period of development, profoundly affects cognitive function. Sodium-potassium adenosine 5'-triphosphatase (Na(+), K(+)-ATPase) is a crucial enzyme responsible for the active transport of sodium and potassium ions in the CNS necessary to maintain the ionic gradient for neuronal excitability. Studies suggest that Na(+), K(+)-ATPase might play a role on memory formation. Moreover, THs were proposed to stimulate Na(+), K(+)-ATPase activity in the heart of some species. In this work we investigated the effect of a chronic administration of L-thyroxine (L-T4) or propylthiouracil (PTU), an antithyroid drug, on some behavioral paradigms: inhibitory avoidance task, open field task, plus maze and Y-maze, and on the activity of Na(+), K(+)-ATPase in the rat parietal cortex and hippocampus. By using treatments which have shown to induce alterations in THs levels similar to those found in hyperthyroid and hypothyroid patients, we aimed to understand the effect of an altered hyperthyroid and hypothyroid state on learning and memory and on the activity of Na(+), K(+)-ATPase. Our results showed that a hyper and hypothyroid state can alter animal behavior and they also might indicate an effect of THs on learning and memory.


Subject(s)
Brain/drug effects , Memory/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Thyroid Hormones/pharmacology , Analysis of Variance , Animals , Antimetabolites/pharmacology , Behavior, Animal , Brain/cytology , Brain/enzymology , Drug Administration Schedule , Male , Maze Learning/drug effects , Propylthiouracil/pharmacology , Rats , Rats, Wistar , Reaction Time/drug effects , Statistics, Nonparametric , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...