Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Arch Biochem Biophys ; 749: 109792, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37863349

RESUMO

Phenylketonuria (PKU) is the most common inherited metabolic disorders caused by severe deficiency or absence of phenylalanine hydroxylase activity that converts phenylalanine (Phe) to tyrosine. PKU patients were treated with a Phe restricted diet supplemented with a special formula containing l-carnitine (L-car), well-known antioxidant compound. The lack of treatment can cause neurological and cognitive impairment, as severe mental retardation, neuronal cell loss and synaptic density reduction. Although Phe has been widely demonstrated to be involved in PKU neurotoxicity, the mechanisms responsible for the CNS injury are still not fully known. In this work, we evaluated markers of neurodegeneration, namely BDNF (brain-derived neurotrophic factor), PAI-1 total (Plasminogen activator inhibitor-1 total), Cathepsin D, PDGF AB/BB (platelet-derived growth factor), and NCAM (neuronal adhesion molecule) in plasma of PKU patients at early and late diagnosis and under treatment. We found decreased Phe levels and increased L-car concentrations in PKU patients treated with L-car compared to the other groups, indicating that the proposed treatment was effective. Furthermore, we found increased BDNF levels in the patients under treatment compared to patients at early diagnosis, and a positive correlation between BDNF and L-car and a negative correlation between BDNF and Phe. Our results may indicate that in PKU patients treated with L-car there is an attempt to adjust neuronal plasticity and recover the damage suffered, reflecting a compensatory response to brain injury.


Assuntos
Carnitina , Fenilcetonúrias , Humanos , Fator Neurotrófico Derivado do Encéfalo , Fenilcetonúrias/tratamento farmacológico , Suplementos Nutricionais , Antioxidantes , Fenilalanina , Becaplermina
2.
Cell Biochem Funct ; 41(4): 490-500, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37170672

RESUMO

Phenylketonuria (PKU) was the first genetic disease to have an effective therapy, which consists of phenylalanine intake restriction. However, there are patients who do not adhere to treatment and/or are not submitted to neonatal screening. PKU patients present L-carnitine (L-car) deficiency, compound that has demonstrated an antioxidant and anti-inflammatory role in metabolic diseases. This study evaluated the effect caused by exposure time to high Phe levels in PKU patients at early and late diagnosis, through pro- and anti-inflammatory cytokines, as well as the L-car effect in patients under treatment. It was observed that there was a decrease in phenylalanine levels in treated patients compared to patients at diagnosis, and an increase in L-car levels in the patients under treatment. Inverse correlation between Phe versus L-car and nitrate plus nitrite versus L-car in PKU patients was also showed. We found increased proinflammatory cytokines levels: interleukin (IL)-1ß, interferons (IFN)-gamma, IL-2, tumor necrosis factor (TNF)-alpha, IL-8 and IL-6 in the patients at late diagnosis compared to controls, and IL-8 in the patients at early diagnosis and treatment compared to controls. Increased IL-2, TNF-alpha, IL-6 levels in the patients at late diagnosis compared to early diagnosis were shown, and reduced IL-6 levels in the treated patients compared to patients at late diagnosis. Moreover, it verified a negative correlation between IFN-gamma and L-car in treated patients. Otherwise, it was observed that there were increased IL-4 levels in the patients at late diagnosis compared to early diagnosis, and reduction in treated patients compared to late diagnosed patients. In urine, there was an increase in 8-isoprostane levels in the patients at diagnosis compared to controls and a decrease in oxidized guanine species in the treated patients compared to the diagnosed patients. Our results demonstrate for the first time in literature that time exposure to high Phe concentrations generates a proinflammatory status, especially in PKU patients with late diagnosis. A pro-oxidant status was verified in not treated PKU patients. Our results demonstrate the importance of early diagnosis and prompt start of treatment, in addition to the importance of L-car supplementation, which can improve cellular defense against inflammation and oxidative damage in PKU patients.


Assuntos
Citocinas , Fenilcetonúrias , Recém-Nascido , Humanos , Fenilalanina , Diagnóstico Tardio , Interleucina-2 , Interleucina-6 , Interleucina-8 , Carnitina/farmacologia , Fenilcetonúrias/diagnóstico , Fenilcetonúrias/tratamento farmacológico , Fenilcetonúrias/urina , Fator de Necrose Tumoral alfa
3.
Metab Brain Dis ; 36(5): 1015-1027, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33620579

RESUMO

Maple syrup urine disease (MSUD) is a genetic disorder that leads the accumulation of branched-chain amino acids (BCAA) leucine (Leu), isoleucine, valine and metabolites. The symptomatology includes psychomotor delay and mental retardation. MSUD therapy comprises a lifelong protein strict diet with low BCAA levels and is well established that high concentrations of Leu and/or its ketoacid are associated with neurological symptoms. Recently, it was demonstrated that the phenylbutyrate (PBA) have the ability to decrease BCAA concentrations. This work aimed the development of lipid-based nanoparticles loaded with PBA, capable of targeting to the central nervous system in order to verify its action mechanisms on oxidative stress and cell death in brain of rats subjected to a MSUD chronic model. PBA-loaded nanoparticles treatment was effective in significantly decreasing BCAA concentration in plasma and Leu in the cerebral cortex of MSUD animals. Furthermore, PBA modulate the activity of catalase, superoxide dismutase, glutathione peroxidase and glutathione reductase enzymes, as well as preventing the oxidative damage to lipid membranes and proteins. PBA was also able to decrease the glial fibrillary acidic protein concentrations and partially decreased the reactive species production and caspase-3 activity in MSUD rats. Taken together, the data indicate that the PBA-loaded nanoparticles could be an efficient adjuvant in the MSUD therapy, protecting against oxidative brain damage and neuroinflammation.


Assuntos
Aminoácidos de Cadeia Ramificada/sangue , Córtex Cerebral/efeitos dos fármacos , Doença da Urina de Xarope de Bordo/metabolismo , Nanopartículas/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Fenilbutiratos/administração & dosagem , Animais , Catalase/metabolismo , Córtex Cerebral/metabolismo , Glutationa Peroxidase/metabolismo , Doença da Urina de Xarope de Bordo/sangue , Doença da Urina de Xarope de Bordo/induzido quimicamente , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo
4.
Cell Mol Neurobiol ; 38(8): 1505-1516, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30302628

RESUMO

X-linked adrenoleukodystrophy (X-ALD) is an inherited neurometabolic disorder caused by disfunction of the ABCD1 gene, which encodes a peroxisomal protein responsible for the transport of the very long-chain fatty acids from the cytosol into the peroxisome, to undergo ß-oxidation. The mainly accumulated saturated fatty acids are hexacosanoic acid (C26:0) and tetracosanoic acid (C24:0) in tissues and body fluids. This peroxisomal disorder occurs in at least 1 out of 20,000 births. Considering that pathophysiology of this disease is not well characterized yet, and glial cells are widely used in studies of protective mechanisms against neuronal oxidative stress, we investigated oxidative damages and inflammatory effects of vesicles containing lecithin and C26:0, as well as the protection conferred by N-acetyl-L-cysteine (NAC), trolox (TRO), and rosuvastatin (RSV) was assessed. It was verified that glial cells exposed to C26:0 presented oxidative DNA damage (measured by comet assay and endonuclease III repair enzyme), enzymatic oxidative imbalance (high catalase activity), nitrative stress [increased nitric oxide (NO) levels], inflammation [high Interleukin-1beta (IL-1ß) levels], and induced lipid peroxidation (increased isoprostane levels) compared to native glial cells without C26:0 exposure. Furthermore, NAC, TRO, and RSV were capable to mitigate some damages caused by the C26:0 in glial cells. The present work yields experimental evidence that inflammation, oxidative, and nitrative stress may be induced by hexacosanoic acid, the main accumulated metabolite in X-ALD, and that antioxidants might be considered as an adjuvant therapy for this severe neurometabolic disease.


Assuntos
Acetilcisteína/farmacologia , Cromanos/farmacologia , Ácidos Graxos/farmacologia , Inflamação/patologia , Neuroglia/patologia , Estresse Nitrosativo , Estresse Oxidativo , Rosuvastatina Cálcica/farmacologia , Animais , Antioxidantes/metabolismo , Catalase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Vesículas Citoplasmáticas/metabolismo , Dano ao DNA , Interleucina-1beta/metabolismo , Isoprostanos/metabolismo , Neuroglia/metabolismo , Fármacos Neuroprotetores/farmacologia , Nitratos/metabolismo , Nitritos/metabolismo , Estresse Nitrosativo/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos
5.
Metab Brain Dis ; 33(1): 333-342, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29260360

RESUMO

Several studies have examined neonatal diabetes, a rare disease characterized by hyperglycemia and low insulin levels that is usually diagnosed in the first 6 month of life. Recently, the effects of diabetes on the brain have received considerable attention. In addition, hyperglycemia may perturb brain function and might be associated with neuronal death in adult rats. However, few studies have investigated the damaging effects of neonatal hyperglycemia on the rat brain during central nervous system (CNS) development, particularly the mechanisms involved in the disease. Thus, in the present work, we investigated whether neonatal hyperglycemia induced by streptozotocin (STZ) promoted cell death and altered the levels of proteins involved in survival/death pathways in the rat brain. Cell death was assessed using FluoroJade C (FJC) staining and the expression of the p38 mitogen-activated protein kinase (p38), phosphorylated-c-Jun amino-terminal kinase (p-JNK), c-Jun amino-terminal kinase (JNK), protein kinase B (Akt), phosphorylated-protein kinase B (p-Akt), glycogen synthase kinase-3ß (Gsk3ß), B-cell lymphoma 2 (Bcl2) and Bcl2-associated X protein (Bax) protein were measured by Western blotting. The main results of this study showed that the metabolic alterations observed in diabetic rats (hyperglycemia and hypoinsulinemia) increased p38 expression and decreased p-Akt expression, suggesting that cell survival was altered and cell death was induced, which was confirmed by FJC staining. Therefore, the metabolic conditions observed during neonatal hyperglycemia may contribute to the harmful effect of diabetes on the CNS in a crucial phase of postnatal neuronal development.


Assuntos
Encéfalo/patologia , Morte Celular/fisiologia , Hiperglicemia/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Encéfalo/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Feminino , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Neurônios/metabolismo , Fosforilação , Ratos Wistar , Proteína X Associada a bcl-2/metabolismo
6.
Biochim Biophys Acta ; 1862(9): 1608-16, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27251652

RESUMO

Mucopolysaccharidosis type II (MPS II) is a lysosomal storage disease caused by a deficient activity of iduronate-2-sulfatase, leading to abnormal accumulation of glycosaminoglycans (GAG). The main treatment for MPS II is enzyme replacement therapy (ERT). Previous studies described potential benefits of six months of ERT against oxidative stress in patients. Thus, the aim of this study was to investigate oxidative, nitrative and inflammatory biomarkers in MPS II patients submitted to long term ERT. It were analyzed urine and blood samples from patients on ERT (mean time: 5.2years) and healthy controls. Patients presented increased levels of lipid peroxidation, assessed by urinary 15-F2t-isoprostane and plasmatic thiobarbituric acid-reactive substances. Concerning to protein damage, urinary di-tyrosine (di-Tyr) was increased in patients; however, sulfhydryl and carbonyl groups in plasma were not altered. It were also verified increased levels of urinary nitrate+nitrite and plasmatic nitric oxide (NO) in MPS II patients. Pro-inflammatory cytokines IL-1ß and TNF-α were increased in treated patients. GAG levels were correlated to di-Tyr and nitrate+nitrite. Furthermore, IL-1ß was positively correlated with TNF-α and NO. Contrastingly, we did not observed alterations in erythrocyte superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase activities, in reduced glutathione content and in the plasmatic antioxidant capacity. Although some parameters were still altered in MPS II patients, these results may suggest a protective role of long-term ERT against oxidative stress, especially upon oxidative damage to protein and enzymatic and non-enzymatic defenses. Moreover, the redox imbalance observed in treated patients seems to be GAG- and pro-inflammatory cytokine-related.


Assuntos
Citocinas/metabolismo , Terapia de Reposição de Enzimas , Glicosaminoglicanos/metabolismo , Mucopolissacaridose II/tratamento farmacológico , Mucopolissacaridose II/metabolismo , Adolescente , Adulto , Estudos de Casos e Controles , Criança , Humanos , Iduronato Sulfatase/uso terapêutico , Interleucina-1beta/metabolismo , Masculino , Mucopolissacaridose II/imunologia , Estresse Nitrosativo/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Adulto Jovem
7.
Cell Mol Neurobiol ; 35(6): 899-911, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25805165

RESUMO

Cystathionine-ß-synthase (CBS) deficiency is the main cause of homocystinuria. Homocysteine (Hcy), methionine, and other metabolites of Hcy accumulate in the body of affected patients. Despite the fact that thromboembolism represents the major cause of morbidity in CBS-deficient patients, the mechanisms of cardiovascular alterations found in homocystinuria remain unclear. In this work, we evaluated the lipid and inflammatory profile, oxidative protein damage, and the activities of the enzymes paraoxonase (PON1) and butyrylcholinesterase (BuChE) in plasma of CBS-deficient patients at diagnosis and during the treatment (protein-restricted diet supplemented with pyridoxine, folic acid, betaine, and vitamin B12). We also investigated the effect of folic acid and vitamin B12 on these parameters. We found a significant decrease in HDL cholesterol and apolipoprotein A1 (ApoA-1) levels, as well as in PON1 activity in both untreated and treated CBS-deficient patients when compared to controls. BuChE activity and IL-6 levels were significantly increased in not treated patients. Furthermore, significant positive correlations between PON1 activity and sulphydryl groups and between IL-6 levels and carbonyl content were verified. Moreover, vitamin B12 was positively correlated with PON1 and ApoA-1 levels, while folic acid was inversely correlated with total Hcy concentration, demonstrating the importance of this treatment. Our results also demonstrated that CBS-deficient patients presented important alterations in biochemical parameters, possibly caused by the metabolites of Hcy, as well as by oxidative stress, and that the adequate adherence to the treatment is essential to revert or prevent these alterations.


Assuntos
Arildialquilfosfatase/sangue , Butirilcolinesterase/sangue , Homocistinúria/sangue , Lipídeos/sangue , Oxidantes/sangue , Adolescente , Adulto , Estudos de Casos e Controles , Criança , Pré-Escolar , Cistationina beta-Sintase/deficiência , Cistationina beta-Sintase/genética , Feminino , Ácido Fólico/sangue , Ácido Fólico/fisiologia , Homocistinúria/genética , Humanos , Masculino , Estresse Oxidativo/fisiologia , Vitamina B 12/sangue , Vitamina B 12/fisiologia , Adulto Jovem
8.
Mol Cell Biochem ; 403(1-2): 159-67, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25682169

RESUMO

Recently, the consequences of diabetes on the central nervous system (CNS) have received great attention. However, the mechanisms by which hyperglycemia affects the central nervous system remain poorly understood. In addition, recent studies have shown that hyperglycemia induces oxidative damage in the adult rat brain. In this regard, no study has assessed oxidative stress as a possible mechanism that affects the brain normal function in neonatal hyperglycemic rats. Thus, the present study aimed to investigate whether neonatal hyperglycemia elicits oxidative stress in the brain of neonate rats subjected to a streptozotocin-induced neonatal hyperglycemia model (5-day-old rats). The activities of glucose-6-phosphate-dehydrogenase (G6PD), 6-phosphogluconate-dehydrogenase (6-PGD), NADPH oxidase (Nox), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSHPx), the production of superoxide anion, the thiobarbituric acid-reactive substances (TBA-RS), and the protein carbonyl content were measured. Neonatal hyperglycemic rats presented increased activities of G6PD, 6PGD, and Nox, which altogether may be responsible for the enhanced production of superoxide radical anion that was observed. The enhanced antioxidant enzyme activities (SOD, CAT, and GSHPx) that were observed in neonatal hyperglycemic rats, which may be caused by a rebound effect of oxidative stress, were not able to hinder the observed lipid peroxidation (TBA-RS) and protein damage in the brain. Consequently, these results suggest that oxidative stress could represent a mechanism that explains the harmful effects of neonatal hyperglycemia on the CNS.


Assuntos
Encéfalo/enzimologia , Encéfalo/patologia , Hiperglicemia/patologia , NADPH Oxidases/metabolismo , Estresse Oxidativo , Via de Pentose Fosfato , Animais , Animais Recém-Nascidos , Catalase/metabolismo , Glutationa Peroxidase/metabolismo , Carbonilação Proteica , Ratos Wistar , Superóxidos/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
9.
Metab Brain Dis ; 30(5): 1167-74, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26002427

RESUMO

Maple Syrup Urine Disease (MSUD) is a metabolic disorder caused by a severe deficiency of the branched-chain α-keto acid dehydrogenase complex activity which leads to the accumulation of branched-chain amino acids (BCAA) leucine (Leu), isoleucine and valine and their respective α-keto-acids in body fluids. The main symptomatology presented by MSUD patients includes ketoacidosis, failure to thrive, poor feeding, apnea, ataxia, seizures, coma, psychomotor delay and mental retardation, but, the neurological pathophysiologic mechanisms are poorly understood. The treatment consists of a low protein diet and a semi-synthetic formula restricted in BCAA and supplemented with essential amino acids. It was verified that MSUD patients present L-carnitine (L-car) deficiency and this compound has demonstrated an antioxidant and anti-inflammatory role in metabolic diseases. Since there are no studies in the literature reporting the inflammatory profile of MSUD patients and the L-car role on the inflammatory response in this disorder, the present study evaluates the effect of L-car supplementation on plasma inflammatory cytokines interleukin-1ß (IL-1ß), interleukin-6 (IL-6), interferon-gamma (INF-É£), and a correlation with malondialdehyde (MDA), as a marker of oxidative damage, and with free L-car plasma levels in treated MSUD patients. Significant increases of IL-1ß, IL-6, and INF-É£ were observed before the treatment with L-car. Moreover, there is a negative correlation between all cytokines tested and L-car concentrations and a positive correlation among the MDA content and IL-1ß and IL-6 values. Our data show that L-car supplementation can improve cellular defense against inflammation and oxidative stress in MSUD patients and may represent an additional therapeutic approach to the patients affected by this disease.


Assuntos
Carnitina/uso terapêutico , Suplementos Nutricionais , Mediadores da Inflamação/sangue , Doença da Urina de Xarope de Bordo/sangue , Doença da Urina de Xarope de Bordo/tratamento farmacológico , Criança , Pré-Escolar , Feminino , Humanos , Inflamação/sangue , Inflamação/tratamento farmacológico , Masculino
10.
Cell Mol Neurobiol ; 34(2): 157-65, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24220995

RESUMO

Maple syrup urine disease (MSUD) is a metabolic disease caused by a deficiency in the branched-chain α-keto acid dehydrogenase complex, leading to the accumulation of branched-chain keto acids and their corresponding branched-chain amino acids (BCAA) in patients. Treatment involves protein-restricted diet and the supplementation with a specific formula containing essential amino acids (except BCAA) and micronutrients, in order to avoid the appearance of neurological symptoms. Although the accumulation of toxic metabolites is associated to appearance of symptoms, the mechanisms underlying the brain damage in MSUD remain unclear, and new evidence has emerged indicating that oxidative stress contributes to this damage. In this context, this review addresses some of the recent findings obtained from cells lines, animal studies, and from patients indicating that oxidative stress is an important determinant of the pathophysiology of MSUD. Recent works have shown that the metabolites accumulated in the disease induce morphological alterations in C6 glioma cells through nitrogen reactive species generation. In addition, several works demonstrated that the levels of important antioxidants decrease in animal models and also in MSUD patients (what have been attributed to protein-restricted diets). Also, markers of lipid, protein, and DNA oxidative damage have been reported in MSUD, probably secondary to the high production of free radicals. Considering these findings, it is well-established that oxidative stress contributes to brain damage in MSUD, and this review offers new perspectives for the prevention of the neurological damage in MSUD, which may include the use of appropriate antioxidants as a novel adjuvant therapy for patients.


Assuntos
Doença da Urina de Xarope de Bordo/patologia , Sistema Nervoso/patologia , Estresse Oxidativo , Animais , Antioxidantes/metabolismo , Modelos Animais de Doenças , Radicais Livres/metabolismo , Humanos
11.
J Inherit Metab Dis ; 37(5): 783-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24623196

RESUMO

The objective of this study was to test whether macromolecule oxidative damage and altered enzymatic antioxidative defenses occur in patients with medium-chain acyl coenzyme A dehydrogenase (MCAD) deficiency. We performed a cross-sectional observational study of in vivo parameters of lipid and protein oxidative damage and antioxidant defenses in asymptomatic, nonstressed, MCAD-deficient patients and healthy controls. Patients were subdivided into three groups based on therapy: patients without prescribed supplementation, patients with carnitine supplementation, and patients with carnitine plus riboflavin supplementation. Compared with healthy controls, nonsupplemented MCAD-deficient patients and patients receiving carnitine supplementation displayed decreased plasma sulfhydryl content (indicating protein oxidative damage). Increased erythrocyte superoxide dismutase (SOD) activity in patients receiving carnitine supplementation probably reflects a compensatory mechanism for scavenging reactive species formation. The combination of carnitine plus riboflavin was not associated with oxidative damage. These are the first indications that MCAD-deficient patients experience protein oxidative damage and that combined supplementation of carnitine and riboflavin may prevent these biochemical alterations. Results suggest involvement of free radicals in the pathophysiology of MCAD deficiency. The underlying mechanisms behind the increased SOD activity upon carnitine supplementation need to be determined. Further studies are necessary to determine the clinical relevance of oxidative stress, including the possibility of antioxidant therapy.


Assuntos
Acil-CoA Desidrogenase/deficiência , Antioxidantes/metabolismo , Erros Inatos do Metabolismo Lipídico/metabolismo , Estresse Oxidativo , Proteínas/metabolismo , Acil-CoA Desidrogenase/metabolismo , Adolescente , Adulto , Carnitina/uso terapêutico , Criança , Pré-Escolar , Estudos Transversais , Eritrócitos/metabolismo , Feminino , Humanos , Lactente , Recém-Nascido , Metabolismo dos Lipídeos/genética , Masculino , Riboflavina/uso terapêutico , Vitaminas/uso terapêutico , Adulto Jovem
12.
Cell Biochem Funct ; 32(8): 711-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25431174

RESUMO

There is increasing evidence suggesting that oxidative stress plays an important role in the development of many chronic and degenerative conditions such as diabetic encephalopathy and depression. Considering that diabetic rats and mice present higher depressive-like behaviour when submitted to the forced swimming test and that treatment with insulin and/or clonazepam is able to reverse the behavioural changes of the diabetic rats, the present work investigated the antioxidant status, specifically total antioxidant reactivity and antioxidant potential of insulin and clonazepam, as well as the effect of this drugs upon protein oxidative damage and reactive species formation in cortex, hippocampus and striatum from diabetic rats submitted to forced swimming test. It was verified that longer immobility time in diabetic rats and insulin plus clonazepam treatment reversed this depressive-like behaviour. Moreover, data obtained in this study allowed to demonstrate through different parameters such as protein carbonyl content, 2'7'-dichlorofluorescein oxidation, catalase, superoxide dismutase, glutathione peroxidase assay, total radical-trapping antioxidant potential and total antioxidant reactivity that there is oxidative stress in cortex, hippocampus and striatum from diabetic rats under depressive-like behaviour and highlight the insulin and/or clonazepam effect in these different brain areas, restoring antioxidant status and protein damage.


Assuntos
Anticonvulsivantes/uso terapêutico , Encefalopatias/complicações , Clonazepam/uso terapêutico , Depressão/tratamento farmacológico , Diabetes Mellitus Experimental/complicações , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Animais , Antioxidantes/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Depressão/etiologia , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
13.
Metab Brain Dis ; 28(4): 563-70, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23532706

RESUMO

Diabetes mellitus is characterized by hyperglycemia resulting from defects on insulin secretion, insulin action, or both. It has recently become clear that the central nervous system is not spared from the deleterious effects of diabetes, since diabetic encephalopathy was recognized as a complication of this heterogeneous metabolic disorder. There is a well recognized association between depression and diabetes, once prevalence of depression in diabetic patients is higher than in general population, and clonazepam is being used to treat this complication. Oxidative stress is widely accepted as playing a key mediatory role in the development and progression of diabetes and its complications. In this work we analyzed DNA damage by comet assay and lipid damage in prefrontal cortex, hippocampus and striatum of streptozotocin-induced diabetic rats submitted to the forced swimming test. It was verified that the diabetic group presented DNA and lipid damage in the brain areas evaluated, when compared to the control groups. Additionally, a significant reduction of the DNA and lipid damage in animals treated with insulin and/or clonazepam was observed. These data suggest that the association of these two drugs could protect against DNA and lipid damage in diabetic rats submitted to the forced swimming test, an animal model of depression.


Assuntos
Encéfalo/efeitos dos fármacos , Clonazepam/uso terapêutico , Depressão/tratamento farmacológico , Diabetes Mellitus Experimental/tratamento farmacológico , Moduladores GABAérgicos/uso terapêutico , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Clonazepam/farmacologia , Dano ao DNA/efeitos dos fármacos , Depressão/complicações , Depressão/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Moduladores GABAérgicos/farmacologia , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Masculino , Malondialdeído/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar
14.
Pharm Biol ; 51(5): 533-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23368939

RESUMO

CONTEXT: It is known that oxidative stress occurs in peripheral blood in an experimental animal model of diabetes and depression, and acute treatment with insulin and clonazepam (CNZ) has a protective effect on oxidative stress in this model. OBJECTIVE: This study evaluated the effect of insulin plus CNZ on oxidative stress parameters in the liver of diabetic male rats induced with streptozotocin (STZ) and subjected to forced swimming test (FST). MATERIALS AND METHODS: Diabetes was induced by a single intraperitoneal (i.p.) dose of STZ 60 mg/kg in male Wistar rats. Insulin (4 IU/kg) plus CNZ acute i.p. treatment (0.25 mg/kg) was administered 24, 5 and 1 h before the FST. Nondiabetic control rats received i.p. injections of saline (1 mL/kg). Protein oxidative damage was evaluated by carbonyl formation and the antioxidant redox parameters were analyzed by the measurements of enzymatic activities of the superoxide dismutase (SOD), catalase and glyoxalase I (GLO). Glycemia levels also were determined. RESULTS: Our present study has shown an increase in carbonyl content from diabetic rats subjected to FST (2.04 ± 0.55), while the activity of catalase (51.83 ± 19.02) and SOD (2.30 ± 1.23) were significantly decreased in liver from these animals, which were reverted by the treatment. Also, the activity of GLO (0.15 ± 0.02) in the liver of the animals was decreased. DISCUSSION AND CONCLUSION: Our findings showed that insulin plus CNZ acute treatment ameliorate the antioxidant redox parameters and protect against protein oxidative damage in the liver of diabetic rats subjected to FST.


Assuntos
Clonazepam/farmacologia , Depressão/tratamento farmacológico , Diabetes Mellitus Experimental/tratamento farmacológico , Insulina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Catalase/metabolismo , Clonazepam/administração & dosagem , Depressão/fisiopatologia , Diabetes Mellitus Experimental/fisiopatologia , Modelos Animais de Doenças , Quimioterapia Combinada , Insulina/administração & dosagem , Lactoilglutationa Liase/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Oxirredução/efeitos dos fármacos , Carbonilação Proteica/efeitos dos fármacos , Ratos , Ratos Wistar , Estreptozocina , Superóxido Dismutase/metabolismo , Fatores de Tempo
15.
Mol Genet Metab ; 106(2): 231-6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22525090

RESUMO

Oxidative stress plays an important role in the pathophysiology of neurodegenerative diseases, including X-linked adrenoleukodystrophy (X-ALD). In the present work, we evaluated lipid (malondialdehyde [MDA] content) and protein (sulfhydryl and carbonyl contents) oxidative damage parameters in plasma from X-ALD patients before and after bone marrow transplant (BMT), in order to verify if this treatment is capable to alter the oxidative parameters studied. We also evaluated the plasma concentration of hexacosanoic acid (C26:0) from X-ALD patients and correlated it with the oxidative damage parameters investigated. We observed that MDA content was significantly increased in plasma of X-ALD patients before BMT and after BMT when compared to controls, and that it was significantly reduced in plasma of X-ALD after BMT when compared to the before BMT group. These results indicate that lipid peroxidation is stimulated in X-ALD patients but there is a significant reduction of lipid peroxidation after BMT. Next, we observed a significant reduction of sulfhydryl content in plasma of X-ALD patients before BMT compared to controls indicating protein oxidative damage and that this measurement was increased in these patients after BMT as compared to before BMT. We found no significant differences in plasma carbonyl content in X-ALD patients before and after BMT as compared to controls. However, we observed a significant reduction in this parameter in X-ALD patients after BMT compared to before BMT. Finally, C26:0 plasma concentration was significantly reduced in X-ALD patients after BMT when compared to before BMT. We found no significant correlations between MDA and carbonyl values with C26:0 levels of the patients before BMT and after BMT, but a significant inverse correlation between sulfhydryl content and C26:0 levels was detected. In conclusion, the present study reinforces the hypothesis that lipid peroxidation and protein damage are induced in plasma of X-ALD patients and, in addition, demonstrates that BMT treatment is capable to reduce this pathogenic process. Taken together, the data obtained from plasma of X-ALD patients before and after BMT showing induction and protection, respectively, of oxidative stress, allowed to suggest that BMT, when well succeeded and under the recommendations, is effective to reduce C26:0 plasma levels and the increased lipid and protein oxidative damage in X-ALD.


Assuntos
Adrenoleucodistrofia/metabolismo , Adrenoleucodistrofia/terapia , Transplante de Medula Óssea , Estresse Oxidativo , Adolescente , Proteínas Sanguíneas/metabolismo , Criança , Pré-Escolar , Humanos , Masculino , Malondialdeído/sangue , Compostos de Sulfidrila/sangue
16.
Cell Mol Neurobiol ; 32(1): 77-82, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21833551

RESUMO

Propionic (PA) and methylmalonic (MMA) acidurias are inherited disorders caused by deficiency of propionyl-CoA carboxylase and methylmalonyl-CoA mutase, respectively. Affected patients present acute metabolic crises in the neonatal period and long-term neurological deficits. Treatments of these diseases include a protein restricted diet and L: -carnitine supplementation. L: -Carnitine is widely used in the therapy of these diseases to prevent secondary L: -carnitine deficiency and promote detoxification, and several recent in vitro and in vivo studies have reported antioxidant and antiperoxidative effects of this compound. In this study, we evaluated the oxidative stress parameters, isoprostane and di-tyrosine levels, and the antioxidant capacity, in urine from patients with PA and MMA at the diagnosis, and during treatment with L: -carnitine and protein-restricted diet. We verified a significant increase of isoprostanes and di-tyrosine, as well as a significant reduction of the antioxidant capacity in urine from these patients at diagnosis, as compared to controls. Furthermore, treated patients presented a marked reduction of isoprostanes and di-tyrosine levels in relation to untreated patients. In addition, patients with higher levels of protein and lipid oxidative damage, determined by di-tyrosine and isoprostanes levels, also presented lower urinary concentrations of total and free L: -carnitine. In conclusion, the present results indicate that treatment with low protein diet and L: -carnitine significantly reduces urinary biomarkers of protein and lipid oxidative damage in patients with disorders of propionate metabolism and that L: -carnitine supplementation may be specially involved in this protection.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/dietoterapia , Erros Inatos do Metabolismo dos Aminoácidos/urina , Carnitina/uso terapêutico , Estresse Oxidativo/fisiologia , Propionatos/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Antioxidantes/análise , Antioxidantes/metabolismo , Carnitina/administração & dosagem , Carnitina/análise , Carnitina/urina , Criança , Pré-Escolar , Dieta com Restrição de Proteínas , Suplementos Nutricionais , Humanos , Lactente , Recém-Nascido , Análise por Pareamento , Ácido Metilmalônico/metabolismo , Ácido Metilmalônico/urina , Estresse Oxidativo/efeitos dos fármacos , Propionatos/urina , Resultado do Tratamento , Tirosina/análise , Tirosina/urina
17.
Metab Brain Dis ; 26(1): 21-8, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21380499

RESUMO

Maple syrup urine disease (MSUD) is an autosomal recessive inborn error of metabolism caused by deficiency of the activity of the mitochondrial enzyme complex branched-chain α-keto acid dehydrogenase (BCKAD) leading to accumulation of the branched-chain amino acids (BCAA) leucine, isoleucine and valine and their corresponding branched-chain α-keto acids. Affected patients present severe brain dysfunction manifested such as ataxia, seizures, coma, psychomotor delay and mental retardation. The mechanisms of brain damage in this disease remain poorly understood. Recent studies have shown that oxidative stress may be involved in neuropathology of MSUD. L-Carnitine (L-Car) is considered a potential antioxidant through its action against peroxidation as a scavenger of reactive oxygen species and by its stabilizing effect of damage to cell membranes. In this study we evaluate the possible neuroprotective in vivo effects of L-Car against pro-oxidative effects of BCAA in cerebral cortex of rats. L-Car prevented lipoperoxidation, measured by thiobarbituric acid-reactive substances, protein damage, measured by sulfhydryl and protein carbonyl content and alteration on catalase and glutathione peroxidase activity in rat cortex from a chemically-induced model of MSUD. Our data clearly show that L-Car may be an efficient antioxidant, protecting against the oxidative stress promoted by BCAA. If the present results are confirmed in MSUD patients, this could represent an additional therapeutic approach to the patients affected by this disease.


Assuntos
Antioxidantes/farmacologia , Carnitina , Córtex Cerebral/química , Peroxidação de Lipídeos/efeitos dos fármacos , Doença da Urina de Xarope de Bordo , Estresse Oxidativo/efeitos dos fármacos , 3-Metil-2-Oxobutanoato Desidrogenase (Lipoamida)/deficiência , Animais , Antioxidantes/metabolismo , Carnitina/metabolismo , Carnitina/farmacologia , Catalase/análise , Catalase/metabolismo , Córtex Cerebral/enzimologia , Modelos Animais de Doenças , Feminino , Glutationa Peroxidase/análise , Glutationa Peroxidase/metabolismo , Humanos , Cetoácidos/metabolismo , Masculino , Doença da Urina de Xarope de Bordo/metabolismo , Doença da Urina de Xarope de Bordo/patologia , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/análise
18.
Metab Brain Dis ; 26(4): 291-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21947687

RESUMO

Phenylketonuria (PKU) is caused by deficiency of phenylalanine hydroxylase, leading to accumulation of phenylalanine and its metabolites. Clinical features of PKU patients include mental retardation, microcephaly, and seizures. Oxidative stress has been found in these patients, and is possibly related to neurophysiopatology of PKU. Regular exercise can leads to adaptation of antioxidant system, improving its capacity to detoxification reactive species. The aim of this study was to verify the effects of regular exercise on oxidative stress parameters in the brain of hyperphenylalaninemic rats. Animals were divided into sedentary (Sed) and exercise (Exe) groups, and subdivided into saline (SAL) and hyperphenylalaninemia (HPA). HPA groups were induced HPA through administration of alpha-methylphenylalanine and phenylalanine for 17 days, while SAL groups (n = 16-20) received saline. Exe groups conducted 2-week aerobic exercise for 20 min/day. At 18th day, animals were killed and the brain was homogenized to determine thiobarbituric acid reactives substances (TBA-RS) content, superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) activities. Soleus muscles were collected to determine glycogen content as a marker of oxidative adaptation. Exe groups showed enhanced glycogen content. HPA condition caused an increase in TBA-RS and SOD, and reduces CAT and GPx. Exercise was able to prevent all changes seen in the HPA group, reaching control values, except for SOD activity. No changes were found in the ExeSAL group compared to SedSAL. Hyperphenylalaninemic rats were more responsive to the benefits provided by regular exercise. Physical training may be an interesting strategy to restore the antioxidant system in HPA.


Assuntos
Química Encefálica/fisiologia , Estresse Oxidativo/fisiologia , Fenilcetonúrias/metabolismo , Condicionamento Físico Animal/fisiologia , Animais , Antioxidantes/metabolismo , Catalase/metabolismo , Modelos Animais de Doenças , Glutationa Peroxidase/metabolismo , Glicogênio/metabolismo , Músculo Esquelético/metabolismo , Fenilalanina/efeitos adversos , Fenilalanina/análogos & derivados , Fenilalanina Hidroxilase/deficiência , Fenilcetonúrias/induzido quimicamente , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
19.
Metab Brain Dis ; 25(2): 145-54, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20431931

RESUMO

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.


Assuntos
Antioxidantes/metabolismo , Encefalopatias Metabólicas Congênitas/induzido quimicamente , Cerebelo/efeitos dos fármacos , Cérebro/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ácido Pirrolidonocarboxílico/toxicidade , Fatores Etários , Animais , Antioxidantes/fisiologia , Encefalopatias Metabólicas Congênitas/metabolismo , Cerebelo/metabolismo , Cérebro/metabolismo , Modelos Animais de Doenças , Glutationa Sintase/deficiência , Peroxidação de Lipídeos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Estresse Oxidativo/fisiologia , Ácido Pirrolidonocarboxílico/metabolismo , Ratos , Ratos Wistar
20.
Metab Brain Dis ; 25(2): 251-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20437087

RESUMO

N-Acetylaspartic acid accumulates in Canavan Disease, a severe inherited neurometabolic disease clinically characterized by severe mental retardation, hypotonia, macrocephaly and generalized tonic and clonic type seizures. Considering that the mechanisms of brain damage in this disease remain poorly understood, in the present study we investigated the in vitro and in vivo effects of N-acetylaspartic acid on the activities of catalase, superoxide dismutase and glutathione peroxidase, as well as on hydrogen peroxide concentration in cerebral cortex of 14-day-old rats. Catalase and glutathione peroxidase activities were significantly inhibited, while hydrogen peroxide concentration was significantly enhanced by N-acetylaspartic acid both in vitro and in vivo. In contrast, superoxide dismutase activity was not altered by N-acetylaspartic acid. Our results clearly show that N-acetylaspartic acid impairs the enzymatic antioxidant defenses in rat brain. This could be involved in the pathophysiological mechanisms responsible for the brain damage observed in patients affected by Canavan Disease.


Assuntos
Antioxidantes/metabolismo , Ácido Aspártico/análogos & derivados , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Catalase/metabolismo , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/metabolismo , Superóxido Dismutase/metabolismo , Animais , Ácido Aspártico/metabolismo , Ácido Aspártico/toxicidade , Ácido Aspártico/urina , Encéfalo/enzimologia , Doença de Canavan/metabolismo , Doença de Canavan/fisiopatologia , Catalase/efeitos dos fármacos , Esquema de Medicação , Feminino , Glutationa Peroxidase/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Ratos , Ratos Wistar , Superóxido Dismutase/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA