Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
J Neurosci Res ; 100(4): 992-1007, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34713466

RESUMEN

Glutaric acidemia type I (GA-I) is an inborn error of metabolism of lysine, hydroxylysine, and tryptophan, caused by glutaryl-CoA-dehydrogenase (GCDH) deficiency, characterized by the buildup of toxic organic acids predominantly in the brain. After acute catabolic states, patients usually develop striatal degeneration, but the mechanisms behind this damage are still unknown. Quinolinic acid (QA), a metabolite of the kynurenine pathway, increases especially during infections/inflammatory processes, and could act synergically with organic acids, contributing to the neurological features of GA-I. The aim of this study was to investigate whether QA increases seizure susceptibility and modifies brain oscillation patterns in an animal model of GA-I, the Gcdh-/- mice taking high-lysine diet (Gcdh-/- -Lys). Therefore, the characteristics of QA-induced seizures and changes in brain oscillatory patterns were evaluated by video-electroencephalography (EEG) analysis recorded in Gcdh-/- -Lys, Gcdh+/+ -Lys, and Gcdh-/- -N (normal diet) animals. We found that the number of seizures per animal was similar for all groups receiving QA, Gcdh-/- -Lys-QA, Gcdh+/+ -Lys-QA, and Gcdh-/- -N-QA. However, severe seizures were observed in the majority of Gcdh-/- -Lys-QA mice (82%), and only in 25% of Gcdh+/+ -Lys-QA and 44% of Gcdh-/- -N-QA mice. All Gcdh-/- -Lys animals developed spontaneous recurrent seizures (SRS), but Gcdh-/- -Lys-QA animals had increased number of SRS, higher mortality rate, and significant predominance of lower frequency oscillations on EEG. Our results suggest that QA plays an important role in the neurological features of GA-I, as Gcdh-/- -Lys mice exhibit increased susceptibility to intrastriatal QA-induced seizures and long-term changes in brain oscillations.


Asunto(s)
Lisina , Ácido Quinolínico , Errores Innatos del Metabolismo de los Aminoácidos , Animales , Encéfalo/metabolismo , Encefalopatías Metabólicas , Modelos Animales de Enfermedad , Glutaril-CoA Deshidrogenasa/deficiencia , Humanos , Lisina/metabolismo , Lisina/farmacología , Ratones , Ratones Noqueados , Ácido Quinolínico/toxicidad , Convulsiones/inducido químicamente , Convulsiones/metabolismo
2.
J Neurochem ; 158(2): 262-281, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33837559

RESUMEN

Tissue accumulation and high urinary excretion of ethylmalonic acid (EMA) are found in ethylmalonic encephalopathy (EE), an inherited disorder associated with cerebral and cerebellar atrophy whose pathogenesis is poorly established. The in vitro and in vivo effects of EMA on bioenergetics and redox homeostasis were investigated in rat cerebellum. For the in vitro studies, cerebellum preparations were exposed to EMA, whereas intracerebellar injection of EMA was used for the in vivo evaluation. EMA reduced state 3 and uncoupled respiration in vitro in succinate-, glutamate-, and malate-supported mitochondria, whereas decreased state 4 respiration was observed using glutamate and malate. Furthermore, mitochondria permeabilization and succinate supplementation diminished the decrease in state 3 with succinate. EMA also inhibited the activity of KGDH, an enzyme necessary for glutamate oxidation, in a mixed manner and augmented mitochondrial efflux of α-ketoglutarate. ATP levels were markedly reduced by EMA, reflecting a severe bioenergetic disruption. Docking simulations also indicated interactions between EMA and KGDH and a competition with glutamate and succinate for their mitochondrial transporters. In vitro findings also showed that EMA decreased mitochondrial membrane potential and Ca2+ retention capacity, and induced swelling in the presence of Ca2+ , which were prevented by cyclosporine A and ADP and ruthenium red, indicating mitochondrial permeability transition (MPT). Moreover, EMA, at high concentrations, mildly increased ROS levels and altered antioxidant defenses in vitro and in vivo. Our data indicate that EMA-induced impairment of glutamate and succinate oxidation and MPT may contribute to the pathogenesis of the cerebellum abnormalities in EE.


Asunto(s)
Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Metabolismo Energético/efectos de los fármacos , Glutamatos/metabolismo , Malonatos/toxicidad , Poro de Transición de la Permeabilidad Mitocondrial , Succinatos/metabolismo , Animales , Ácidos Cetoglutáricos/metabolismo , Malatos/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Simulación del Acoplamiento Molecular , Oxidación-Reducción , Consumo de Oxígeno/efectos de los fármacos , Ratas , Ratas Wistar , Succinatos/farmacología
3.
Hum Mol Genet ; 28(6): 928-941, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30445591

RESUMEN

Very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency is the most common defect of mitochondrial long-chain fatty acid ß-oxidation. Patients present with heterogeneous clinical phenotypes affecting heart, liver and skeletal muscle predominantly. The full pathophysiology of the disease is unclear and patient response to current therapeutic regimens is incomplete. To identify additional cellular alterations and explore more effective therapies, mitochondrial bioenergetics and redox homeostasis were assessed in VLCAD-deficient fibroblasts, and several protective compounds were evaluated. The results revealed cellular and tissue changes, including decreased respiratory chain (RC) function, increased reactive oxygen species (ROS) production and altered mitochondrial function and signaling pathways in a variety of VLCAD-deficient fibroblasts. The mitochondrially enriched electron and free radical scavengers JP4-039 and XJB-5-131 improved RC function and decreased ROS production significantly, suggesting that they are viable candidate compounds to further develop to treat VLCAD-deficient patients.


Asunto(s)
Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Antioxidantes/farmacología , Síndromes Congénitos de Insuficiencia de la Médula Ósea/metabolismo , Transporte de Electrón/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Errores Innatos del Metabolismo Lipídico/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Enfermedades Musculares/metabolismo , Acil-CoA Deshidrogenasa de Cadena Larga/metabolismo , Adenosina Trifosfato/metabolismo , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Síndromes Congénitos de Insuficiencia de la Médula Ósea/etiología , Retículo Endoplásmico/metabolismo , Errores Innatos del Metabolismo Lipídico/etiología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Enfermedades Mitocondriales/etiología , Dinámicas Mitocondriales/efectos de los fármacos , Enfermedades Musculares/etiología , Oxidación-Reducción/efectos de los fármacos , Consumo de Oxígeno , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
4.
J Bioenerg Biomembr ; 53(5): 525-539, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34347214

RESUMEN

S-adenosylmethionine (AdoMet) predominantly accumulates in tissues and biological fluids of patients affected by liver dysmethylating diseases, particularly glycine N-methyltransferase, S-adenosylhomocysteine hydrolase and adenosine kinase deficiencies, as well as in some hepatic mtDNA depletion syndromes, whose pathogenesis of liver dysfunction is still poorly established. Therefore, in the present work, we investigated the effects of S-adenosylmethionine (AdoMet) on mitochondrial functions and redox homeostasis in rat liver. AdoMet decreased mitochondrial membrane potential and Ca2+ retention capacity, and these effects were fully prevented by cyclosporin A and ADP, indicating mitochondrial permeability transition (mPT) induction. It was also verified that the thiol-alkylating agent NEM prevented AdoMet-induced ΔΨm dissipation, implying a role for thiol oxidation in the mPT pore opening. AdoMet also increased ROS production and provoked protein and lipid oxidation. Furthermore, AdoMet reduced GSH levels and the activities of aconitase and α-ketoglutarate dehydrogenase. Free radical scavengers attenuated AdoMet effects on lipid peroxidation and GSH levels, supporting a role of ROS in these effects. It is therefore presumed that disturbance of mitochondrial functions associated with mPT and redox unbalance may represent relevant pathomechanisms of liver damage provoked by AdoMet in disorders in which this metabolite accumulates.


Asunto(s)
Hígado/patología , Proteínas de Transporte de Membrana Mitocondrial/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , S-Adenosilmetionina/efectos adversos , Animales , Masculino , Permeabilidad , Ratas , Ratas Wistar
5.
Neurochem Res ; 41(10): 2619-2626, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27278758

RESUMEN

3-Methylglutaric acid (3MGA) is an organic acid that accumulates in various organic acidemias whose patients present neurodegeneration events in children coursing with metabolic acidurias. Limited evidence describes the toxic mechanisms elicited by 3MGA in the brain. Herein, we explored the effects of 3MGA on different toxic endpoints in synaptosomal and mitochondrial-enriched fractions of adult rat brains to provide novel information on early mechanisms evoked by this metabolite. At 1 and 5 mM concentration, 3MGA increased lipid peroxidation, but decreased mitochondrial function only at 5 mM concentration. Despite less intense effects were obtained at 1 mM concentration, its co-administration with the kynurenine pathway (KP) metabolite and N-methyl-D-aspartate receptor (NMDAr) agonist, quinolinic acid (QUIN, 50 and 100 µM), produced toxic synergism on markers of oxidative stress and mitochondrial function. The toxicity of 3MGA per se (5 mM) was prevented by the cannabinoid receptor agonist WIN55,212-2 and the NMDAr antagonist kynurenic acid (KYNA), suggesting cannabinoid and glutamatergic components in the 3MGA pattern of toxicity. The synergic model (3MGA + QUIN) was also sensitive to KYNA and the antioxidant S-allylcysteine, but not to the nitric oxide synthase inhibitor L-nitroarginine methyl ester. These findings suggest various underlying mechanisms involved in the neurotoxicity of 3MGA that may possibly contribute to the neurodegeneration observed in acidemias.


Asunto(s)
Encéfalo/efectos de los fármacos , Meglutol/análogos & derivados , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Sinaptosomas/efectos de los fármacos , Animales , Antioxidantes/farmacología , Encéfalo/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Meglutol/farmacología , Mitocondrias/metabolismo , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Receptores de Cannabinoides/metabolismo , Sinaptosomas/metabolismo
6.
Metab Brain Dis ; 30(4): 1055-62, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25939283

RESUMEN

3-Hydroxy-3-methylglutaryl-CoA lyase (HL) deficiency is a rare autosomal recessive disorderaffecting the final step of leucine degradation and ketogenesis and biochemically characterized by the predominant accumulation of 3-hydroxy-3-methylglutaric (HMG) and 3-methylglutaric (MGA) acids in biological fluids and tissues of affected patients. Considering that previous studies reported that HMG and MGA have pro oxidant properties, the present study evaluated the ex vivo and in vitro effects of HMG and MGA on frequency and index of DNA damage in cerebral cortex and striatum of young rats. The ex vivo effects of both organic acids on 8-hydroxy-2'-deoxyguanosine (OHdG) levels and their in vitro effects on 2',7'-dichlorofluorescin (DCFH) oxidation and glutathione (GSH) concentrations in rat striatum were also determined. We also investigated the ex vivo effects of both organic acids on 8-hydroxy-2'-deoxyguanosine (OHdG) levels in rat striatum. In the ex vivo experiments, DNA damage was determined in striatum homogenates prepared 30 min after a single intrastriatal administration of HMG or MGA. On the other hand, the in vitro evaluation was performed after an incubation of rat cerebral cortex or striatum homogenates or slices in the presence of HMG or MGA during 1 h at 37 °C. We observed that the intrastriatal administration of HMG and MGA increased the frequency and the index of DNA damage, as well as OHdG staining in rat striatum. We also verified that MGA, but not HMG, increased DNA damage frequency and index in vitro in striatum of rats. In contrast, no alterations were verified in vitro in cerebral cortex. Finally, we found that HMG and MGA increased DCFH oxidation and decreased GSH concentrations in rat striatum. Therefore, it may be presumed that DNA damage provoked by HMG and MGA possibly via reactive species generation is involved, at least in part, in the pathophysiology of brain injury, particularly in the striatum of HL-deficient patients.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Daño del ADN/efectos de los fármacos , Meglutol/análogos & derivados , Meglutol/toxicidad , Animales , Cuerpo Estriado/patología , Daño del ADN/fisiología , Relación Dosis-Respuesta a Droga , Inyecciones Intraventriculares , Masculino , Meglutol/administración & dosificación , Ratas , Ratas Wistar
7.
Biochimie ; 219: 21-32, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37541567

RESUMEN

Non ketotic hyperglycinemia (NKH) is an inborn error of glycine metabolism caused by mutations in the genes encoding glycine cleavage system proteins. Classic NKH has a neonatal onset, and patients present with severe neurodegeneration. Although glycine accumulation has been implicated in NKH pathophysiology, the exact mechanisms underlying the neurological damage and white matter alterations remain unclear. We investigated the effects of glycine in the brain of neonatal rats and MO3.13 oligodendroglial cells. Glycine decreased myelin basic protein (MBP) and myelin-associated glycoprotein (MAG) in the corpus callosum and striatum of rats on post-natal day (PND) 15. Glycine also reduced neuroglycan 2 (NG2) and N-methyl-d-aspartate receptor subunit 1 (NR1) in the cerebral cortex and striatum on PND15. Moreover, glycine reduced striatal glutamate aspartate transporter 1 (GLAST) content and neuronal nucleus (NeuN), and increased glial fibrillary acidic protein (GFAP) on PND15. Glycine also increased DCFH oxidation and malondialdehyde levels and decreased GSH concentrations in the cerebral cortex and striatum on PND6, but not on PND15. Glycine further reduced viability but did not alter DCFH oxidation and GSH levels in MO3.13 cells after 48- and 72-h incubation. These data indicate that impairment of myelin structure and glutamatergic system and induction of oxidative stress are involved in the neuropathophysiology of NKH.


Asunto(s)
Hiperglicinemia no Cetósica , Humanos , Animales , Ratas , Hiperglicinemia no Cetósica/genética , Hiperglicinemia no Cetósica/metabolismo , Glicina , Vaina de Mielina/metabolismo , Oxidación-Reducción , Transmisión Sináptica , Homeostasis
8.
Mol Genet Metab ; 109(2): 144-53, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23611578

RESUMEN

3-Hydroxy-3-methylglutaryl-CoA lyase (HL) deficiency is a genetic disorder biochemically characterized by predominant accumulation of 3-hydroxy-3-methylglutaric (HMG) and 3-methylglutaric (MGA) acids in tissues and biological fluids of affected individuals. Clinically, the patients present neurological symptoms and basal ganglia injury, whose pathomechanisms are partially understood. In the present study, we investigated the ex vivo effects of intrastriatal administration of HMG and MGA on important parameters of oxidative stress in striatum of developing rats. Our results demonstrate that HMG and MGA induce lipid and protein oxidative damage. HMG and MGA also increased 2',7'-dichlorofluorescein oxidation, whereas only HMG elicited nitric oxide production, indicating a role for reactive oxygen (HMG and MGA) and nitrogen (HMG) species in these effects. Regarding the enzymatic antioxidant defenses, both organic acids decreased reduced glutathione concentrations and the activities of superoxide dismutase and glutathione reductase and increased glutathione peroxidase activity. HMG also provoked an increase of catalase activity and a diminution of glucose-6-phosphate dehydrogenase activity. We finally observed that antioxidants fully prevented or attenuated HMG-induced alterations of the oxidative stress parameters, further indicating the participation of reactive species in these effects. We also observed that MK-801, a non-competitive antagonist of the N-methyl-D-aspartate (NMDA) receptor, prevented some of these effects, indicating the involvement of the NMDA receptor in HMG effects. The present data provide solid evidence that oxidative stress is induced in vivo by HMG and MGA in rat striatum and it is presumed that this pathomechanism may explain, at least in part, the cerebral alterations observed in HL deficiency.


Asunto(s)
Acetil-CoA C-Acetiltransferasa/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Ganglios Basales/metabolismo , Meglutol/análogos & derivados , Meglutol/metabolismo , Estrés Oxidativo , Acetil-CoA C-Acetiltransferasa/metabolismo , Animales , Antioxidantes/farmacología , Ganglios Basales/crecimiento & desarrollo , Ganglios Basales/patología , Catalasa/metabolismo , Maleato de Dizocilpina/farmacología , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Masculino , Malondialdehído/metabolismo , Carbonilación Proteica , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Superóxido Dismutasa/metabolismo , Vitamina E/farmacología
9.
Mol Genet Metab ; 108(1): 30-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23218171

RESUMEN

Deficiency of glutaryl-CoA dehydrogenase (GCDH) activity or glutaric aciduria type I (GA I) is an inherited neurometabolic disorder biochemically characterized by predominant accumulation of glutaric acid and 3-hydroxyglutaric acid in the brain and other tissues. Affected patients usually present acute striatum necrosis during encephalopathic crises triggered by metabolic stress situations, as well as chronic leukodystrophy and delayed myelination. Considering that the mechanisms underlying the brain injury in this disease are not yet fully established, in the present study we investigated important parameters of oxidative stress in the brain (cerebral cortex, striatum and hippocampus), liver and heart of 30-day-old GCDH deficient knockout (Gcdh(-/-)) and wild type (WT) mice submitted to a normal lysine (Lys) (0.9% Lys), or high Lys diets (2.8% or 4.7% Lys) for 60 h. It was observed that the dietary supplementation of 2.8% and 4.7% Lys elicited noticeable oxidative stress, as verified by an increase of malondialdehyde concentrations (lipid oxidative damage) and 2-7-dihydrodichlorofluorescein (DCFH) oxidation (free radical production), as well as a decrease of reduced glutathione levels and alteration of various antioxidant enzyme activities (antioxidant defenses) in the cerebral cortex and the striatum, but not in the hippocampus, the liver and the heart of Gcdh(-/-) mice, as compared to WT mice receiving the same diets. Furthermore, alterations of oxidative stress parameters in the cerebral cortex and striatum were more accentuated in symptomatic, as compared to asymptomatic Gcdh(-/-) mice exposed to 4.7% Lys overload. Histopathological studies performed in the cerebral cortex and striatum of these animals exposed to high dietary Lys revealed increased expression of oxidative stress markers despite the absence of significant structural damage. The results indicate that a disruption of redox homeostasis in the cerebral cortex and striatum of young Gcdh(-/-) mice exposed to increased Lys diet may possibly represent an important pathomechanism of brain injury in GA I patients under metabolic stress.


Asunto(s)
Encéfalo/metabolismo , Glutaril-CoA Deshidrogenasa/metabolismo , Homeostasis , Lisina/administración & dosificación , Animales , Suplementos Dietéticos , Glutaril-CoA Deshidrogenasa/genética , Ratones , Ratones Noqueados , Oxidación-Reducción , Estrés Oxidativo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
10.
Biochem Pharmacol ; 209: 115433, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36709926

RESUMEN

Inherited metabolic disorders (IMDs) are genetic disorders that cause a disruption of a specific metabolic pathway leading to biochemical, clinical and pathophysiological sequelae. While the metabolite abnormalities in body fluids and tissues can usually be defined by directed or broad-spectrum metabolomic analysis, the pathophysiology of these changes is often not obvious. Mounting evidence has revealed that secondary mitochondrial dysfunction, mainly oxidative phosphorylation impairment and elevated reactive oxygen species, plays a pivotal role in many disorders. Peroxisomal proliferator-activated receptors (PPARs) consist of a group of nuclear hormone receptors (PPARα, PPARß/δ, and PPARγ) that regulate multiple cellular functions and processes, including response to oxidative stress, inflammation, lipid metabolism, and mitochondrial bioenergetics and biogenesis. In this context, the activation of PPARs has been shown to stimulate oxidative phosphorylation and reduce reactive species levels. Thus, pharmacological treatment with PPAR activators, such as fibrates, has gained much attention in the last 15 years. This review summarizes preclinical (animal models and patient-derived cells) and clinical data on the effect of PPARs in IMDs.


Asunto(s)
Enfermedades Metabólicas , PPAR delta , Animales , PPAR alfa , PPAR gamma , Metabolismo de los Lípidos
11.
Toxicol Lett ; 381: 48-59, 2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-37116597

RESUMEN

Redox homeostasis, mitochondrial functions, and mitochondria-endoplasmic reticulum (ER) communication were evaluated in the striatum of rats after 3-nitropropionic acid (3-NP) administration, a recognized chemical model of Huntington's disease (HD). 3-NP impaired redox homeostasis by increasing malondialdehyde levels at 28 days, decreasing glutathione (GSH) concentrations at 21 and 28 days, and the activities of glutathione peroxidase (GPx), superoxide dismutase (SOD) and glutathione S-transferase at 7, 21, and 28 days, catalase at 21 days, and glutathione reductase at 21 and 28 days. Impairment of mitochondrial respiration at 7 and 28 days after 3-NP administration was also observed, as well as reduced activities of succinate dehydrogenase (SDH) and respiratory chain complexes. 3-NP also impaired mitochondrial dynamics and the interactions between ER and mitochondria and induced ER-stress by increasing the levels of mitofusin-1, and of DRP1, VDAC1, Grp75 and Grp78. Synaptophysin levels were augmented at 7 days but reduced at 28 days after 3-NP injection. Finally, bezafibrate prevented 3-NP-induced alterations of the activities of SOD, GPx, SDH and respiratory chain complexes, DCFH oxidation and on the levels of GSH, VDAC1 and synaptophysin. Mitochondrial dysfunction and synaptic disruption may contribute to the pathophysiology of HD and bezafibrate may be considered as an adjuvant therapy for this disorder.


Asunto(s)
Enfermedad de Huntington , Ratas , Animales , Enfermedad de Huntington/inducido químicamente , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/metabolismo , Ratas Wistar , Bezafibrato/efectos adversos , Bezafibrato/metabolismo , Sinaptofisina/metabolismo , Modelos Químicos , Estrés Oxidativo , Glutatión/metabolismo , Superóxido Dismutasa/metabolismo , Mitocondrias/metabolismo , Propionatos/toxicidad , Nitrocompuestos/toxicidad , Nitrocompuestos/metabolismo
12.
Cells ; 12(12)2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37371027

RESUMEN

Sulfite predominantly accumulates in the brain of patients with isolated sulfite oxidase (ISOD) and molybdenum cofactor (MoCD) deficiencies. Patients present with severe neurological symptoms and basal ganglia alterations, the pathophysiology of which is not fully established. Therapies are ineffective. To elucidate the pathomechanisms of ISOD and MoCD, we investigated the effects of intrastriatal administration of sulfite on myelin structure, neuroinflammation, and oxidative stress in rat striatum. Sulfite administration decreased FluoromyelinTM and myelin basic protein staining, suggesting myelin abnormalities. Sulfite also increased the staining of NG2, a protein marker of oligodendrocyte progenitor cells. In line with this, sulfite also reduced the viability of MO3.13 cells, which express oligodendroglial markers. Furthermore, sulfite altered the expression of interleukin-1ß (IL-1ß), interleukin-6 (IL-6), interleukin-10 (IL-10), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS) and heme oxygenase-1 (HO-1), indicating neuroinflammation and redox homeostasis disturbances. Iba1 staining, another marker of neuroinflammation, was also increased by sulfite. These data suggest that myelin changes and neuroinflammation induced by sulfite contribute to the pathophysiology of ISOD and MoCD. Notably, post-treatment with bezafibrate (BEZ), a pan-PPAR agonist, mitigated alterations in myelin markers and Iba1 staining, and IL-1ß, IL-6, iNOS and HO-1 expression in the striatum. MO3.13 cell viability decrease was further prevented. Moreover, pre-treatment with BEZ also attenuated some effects. These findings show the modulation of PPAR as a potential opportunity for therapeutic intervention in these disorders.


Asunto(s)
Bezafibrato , Receptores Activados del Proliferador del Peroxisoma , Ratas , Animales , Bezafibrato/farmacología , Receptores Activados del Proliferador del Peroxisoma/farmacología , Vaina de Mielina , Enfermedades Neuroinflamatorias , Interleucina-6/farmacología , Estrés Oxidativo , Sulfitos/farmacología
13.
Mol Genet Metab ; 106(1): 31-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22445450

RESUMEN

In the present work we evaluated a variety of indicators of oxidative stress in distinct brain regions (striatum, cerebral cortex and hippocampus), the liver, and heart of 30-day-old glutaryl-CoA dehydrogenase deficient (Gcdh(-/-)) mice. The parameters evaluated included thiobarbituric acid-reactive substances (TBA-RS), 2-7-dihydrodichlorofluorescein (DCFH) oxidation, sulfhydryl content, and reduced glutathione (GSH) concentrations. We also measured the activities of the antioxidant enzymes glutathione peroxidase (GPx), glutathione reductase (GR), catalase (CAT), superoxide dismutase (SOD) and glucose-6-phosphate dehydrogenase (G6PD). Under basal conditions glutaric (GA) and 3-OH-glutaric (3OHGA) acids were elevated in all tissues of the Gcdh(-/-) mice, but were essentially absent in WT animals. In contrast there were no differences between WT and Gcdh(-/-) mice in any of the indicators or oxidative stress under basal conditions. Following a single intra-peritoneal (IP) injection of lysine (Lys) there was a moderate increase of brain GA concentration in Gcdh(-/-) mice, but no change in WT. Lys injection had no effect on brain 3OHGA in either WT or Gcdh(-/-) mice. The levels of GA and 3OHGA were approximately 40% higher in striatum compared to cerebral cortex in Lys-treated mice. In the striatum, Lys administration provoked a marked increase of lipid peroxidation, DCFH oxidation, SOD and GR activities, as well as significant reductions of GSH levels and GPx activity, with no alteration of sulfhydryl content, CAT and G6PD activities. There was also evidence of increased lipid peroxidation and SOD activity in the cerebral cortex, along with a decrease of GSH levels, but to a lesser extent than in the striatum. In the hippocampus only mild increases of SOD activity and DCFH oxidation were observed. In contrast, Lys injection had no effect on any of the parameters of oxidative stress in the liver or heart of Gcdh(-/-) or WT animals. These results indicate that in Gcdh(-/-) mice cerebral tissue, particularly the striatum, is at greater risk for oxidative stress than peripheral tissues following Lys administration.


Asunto(s)
Antioxidantes/metabolismo , Encéfalo/metabolismo , Glutaril-CoA Deshidrogenasa , Lisina/administración & dosificación , Estrés Oxidativo , Animales , Encéfalo/enzimología , Glutaril-CoA Deshidrogenasa/deficiencia , Glutaril-CoA Deshidrogenasa/genética , Peroxidación de Lípido , Hígado/enzimología , Hígado/metabolismo , Lisina/efectos adversos , Ratones , Ratones Transgénicos , Miocardio/enzimología , Miocardio/metabolismo , Distribución Tisular
14.
Mol Genet Metab ; 107(3): 375-82, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22999741

RESUMEN

Mitochondrial dysfunction has been proposed to play an important role in the neuropathology of glutaric acidemia type I (GA I). However, the relevance of bioenergetics disruption and the exact mechanisms responsible for the cortical leukodystrophy and the striatum degeneration presented by GA I patients are not yet fully understood. Therefore, in the present work we measured the respiratory chain complexes activities I-IV, mitochondrial respiratory parameters state 3, state 4, the respiratory control ratio and dinitrophenol (DNP)-stimulated respiration (uncoupled state), as well as the activities of α-ketoglutarate dehydrogenase (α-KGDH), creatine kinase (CK) and Na+, K+-ATPase in cerebral cortex, striatum and hippocampus from 30-day-old Gcdh-/- and wild type (WT) mice fed with a normal or a high Lys (4.7%) diet. When a baseline (0.9% Lys) diet was given, we verified mild alterations of the activities of some respiratory chain complexes in cerebral cortex and hippocampus, but not in striatum from Gcdh-/- mice as compared to WT animals. Furthermore, the mitochondrial respiratory parameters and the activities of α-KGDH and CK were not modified in all brain structures from Gcdh-/- mice. In contrast, we found a significant reduction of Na(+), K(+)-ATPase activity associated with a lower degree of its expression in cerebral cortex from Gcdh-/- mice. Furthermore, a high Lys (4.7%) diet did not accentuate the biochemical alterations observed in Gcdh-/- mice fed with a normal diet. Since Na(+), K(+)-ATPase activity is required for cell volume regulation and to maintain the membrane potential necessary for a normal neurotransmission, it is presumed that reduction of this enzyme activity may represent a potential underlying mechanism involved in the brain swelling and cortical abnormalities (cortical atrophy with leukodystrophy) observed in patients affected by GA I.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/patología , Encefalopatías Metabólicas/genética , Encefalopatías Metabólicas/patología , Corteza Cerebral/patología , Cuerpo Estriado/patología , Glutaril-CoA Deshidrogenasa/deficiencia , Hipocampo/patología , ATPasa Intercambiadora de Sodio-Potasio/genética , Errores Innatos del Metabolismo de los Aminoácidos/enzimología , Animales , Encefalopatías Metabólicas/enzimología , Corteza Cerebral/enzimología , Cuerpo Estriado/enzimología , Creatina Quinasa/genética , Creatina Quinasa/metabolismo , Regulación hacia Abajo , Transporte de Electrón/genética , Alimentos Formulados , Expresión Génica , Glutaril-CoA Deshidrogenasa/genética , Hipocampo/enzimología , Humanos , Complejo Cetoglutarato Deshidrogenasa/genética , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Fosforilación Oxidativa , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
15.
Mol Genet Metab ; 107(1-2): 81-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22578804

RESUMEN

Glutaric acidemia type I (GA I) is an inherited neurometabolic disorder caused by a severe deficiency of the mitochondrial glutaryl-CoA dehydrogenase activity leading to accumulation of predominantly glutaric (GA) and 3-hydroxyglutaric (3HGA) acids in the brain and other tissues. Affected patients usually present with hypotonia and brain damage and acute encephalopathic episodes whose pathophysiology is not yet fully established. In this study we investigated important parameters of cellular bioenergetics in brain, heart and skeletal muscle from 15-day-old glutaryl-CoA dehydrogenase deficient mice (Gcdh(-/-)) submitted to a single intra-peritoneal injection of saline (Sal) or lysine (Lys - 8 µmol/g) as compared to wild type (WT) mice. We evaluated the activities of the respiratory chain complexes II, II-III and IV, α-ketoglutarate dehydrogenase (α-KGDH), creatine kinase (CK) and synaptic Na(+), K(+)-ATPase. No differences of all evaluated parameters were detected in the Gcdh(-/-) relatively to the WT mice injected at baseline (Sal). Furthermore, mild increases of the activities of some respiratory chain complexes (II-III and IV) were observed in heart and skeletal muscle of Gcdh(-/-) and WT mice after Lys administration. However, the most marked effects provoked by Lys administration were marked decreases of the activities of Na(+), K(+)-ATPase in brain and CK in brain and skeletal muscle of Gcdh(-/-) mice. In contrast, brain α-KGDH activity was not altered in WT and Gcdh(-/-) injected with Sal or Lys. Our results demonstrate that reduction of Na(+), K(+)-ATPase and CK activities may play an important role in the pathogenesis of the neurodegenerative changes in GA I.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Encefalopatías Metabólicas/metabolismo , Creatina Quinasa/metabolismo , Lisina/farmacología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encefalopatías Metabólicas/tratamiento farmacológico , Modelos Animales de Enfermedad , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/fisiología , Glutaril-CoA Deshidrogenasa/deficiencia , Glutaril-CoA Deshidrogenasa/metabolismo , Corazón/efectos de los fármacos , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Lisina/administración & dosificación , Ratones , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Miocardio/metabolismo
16.
Metab Brain Dis ; 27(4): 521-30, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22798168

RESUMEN

Ornithine, ammonia and homocitrulline are the major metabolites accumulating in hyperornithinemia-hyperammonemia-homocitrullinuria syndrome, a genetic disorder characterized by neurological regression whose pathogenesis is still not understood. The present work investigated the in vivo effects of intracerebroventricular administration of ornithine and homocitrulline in the presence or absence of hyperammonemia induced by intraperitoneal urease treatment on a large spectrum of oxidative stress parameters in cerebral cortex from young rats in order to better understand the role of these metabolites on brain damage. Ornithine increased thiobarbituric acid-reactive substances (TBA-RS) levels and carbonyl formation and decreased total antioxidant status (TAS) levels. We also observed that the combination of hyperammonemia with ornithine resulted in significant decreases of sulfhydryl levels, reduced glutathione (GSH) concentrations and the activities of catalase (CAT) and glutathione peroxidase (GPx), highlighting a synergistic effect of ornithine and ammonia. Furthermore, homocitrulline caused increases of TBA-RS values and carbonyl formation, as well as decreases of GSH concentrations and GPx activity. Hcit with hyperammonemia (urease treatment) decreased TAS and CAT activity. We also showed that urease treatment per se was able to enhance TBA-RS levels. Finally, nitric oxide production was not altered by Orn and Hcit alone or in combination with hyperammonemia. Our data indicate that the major metabolites accumulating in hyperornithinemia-hyperammonemia-homocitrullinuria syndrome provoke lipid and protein oxidative damage and a reduction of the antioxidant defenses in the brain. Therefore, it is presumed that oxidative stress may represent a relevant pathomechanism involved in the brain damage found in patients affected by this disease.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Amoníaco/metabolismo , Encéfalo/metabolismo , Citrulina/análogos & derivados , Homeostasis/fisiología , Ornitina/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/sangre , Errores Innatos del Metabolismo de los Aminoácidos/inducido químicamente , Amoníaco/sangre , Animales , Antioxidantes/metabolismo , Catalasa/metabolismo , Citrulina/metabolismo , Citrulina/orina , Glutatión Peroxidasa/metabolismo , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Masculino , Óxido Nítrico/metabolismo , Ornitina/sangre , Ornitina/toxicidad , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Carbonilación Proteica/efectos de los fármacos , Ratas , Ratas Wistar , Compuestos de Sulfhidrilo/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Ureasa
17.
Mol Neurobiol ; 59(8): 4839-4853, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35639256

RESUMEN

Patients with glutaric aciduria type 1 (GA1), a neurometabolic disorder caused by deficiency of glutaryl-CoA dehydrogenase (GCDH) activity, commonly manifest acute encephalopathy associated with severe striatum degeneration and progressive cortical and striatal injury whose pathogenesis is still poorly known. We evaluated redox homeostasis, inflammatory response, mitochondrial biogenesis and dynamics, endoplasmic reticulum (ER)-mitochondria crosstalk, and ER stress in the brain of GCDH-deficient (Gcdh-/-) and wild-type (Gcdh+/+) mice fed a high Lys chow, which better mimics the human neuropathology mainly characterized by striatal lesions. Increased lipid peroxidation and altered antioxidant defenses, including decreased concentrations of reduced glutathione and increased activities of superoxide dismutase, catalase, and glutathione transferase, were observed in the striatum and cerebral cortex of Gcdh-/- mice. Augmented Iba-1 staining was also found in the dorsal striatum and neocortex, whereas the nuclear content of NF-κB was increased, and the cytosolic content of IκBα decreased in the striatum of the mutant animals, indicating a pro-inflammatory response. Noteworthy, in vivo treatment with the pan-PPAR agonist bezafibrate normalized these alterations. It was also observed that the ER-mitochondria crosstalk proteins VDAC1 and IP3R were reduced, whereas the ER stress protein DDIT3 was augmented in Gcdh-/- striatum, signaling disturbances of these processes. Finally, DRP1 content was elevated in the striatum of Gcdh-/- mice, indicating activated mitochondrial fission. We presume that some of these novel pathomechanisms may be involved in GA1 neuropathology and that bezafibrate should be tested as a potential adjuvant therapy for GA1.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Fármacos Neuroprotectores , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Animales , Bezafibrato/farmacología , Encéfalo/metabolismo , Encefalopatías Metabólicas , Retículo Endoplásmico/metabolismo , Glutaril-CoA Deshidrogenasa/deficiencia , Homeostasis , Humanos , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Oxidación-Reducción
18.
Mol Genet Metab Rep ; 33: 100932, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36338154

RESUMEN

Autoimmune Disease, Multisystem, with Facial Dysmorphism (ADMFD) is an autosomal recessive disorder due to pathogenic variants in the ITCH gene. It is characterized by failure to thrive, dysmorphic facial features, developmental delay, and systemic autoimmunity that can manifest variably with autoimmune hepatitis, thyroiditis, and enteropathy, among other organ manifestations. It was originally described in 10 consanguineous Old Order Amish patients, and more recently in two patients of White British and Black German ethnicities. While the role of ITCH protein in apoptosis and inflammation has previously been characterized, a defect in cellular bioenergetics has not yet been reported in ITCH deficiency. Here we present a Caucasian female originally evaluated for possible mitochondrial respiratory chain deficiency, who ultimately was found to have two novel variants in ITCH with absence of ITCH protein in patient derived fibroblasts. Clinical studies of patient muscle showed mitochondrial DNA copy number of 57% compared to controls. Functional studies in skin fibroblasts revealed decreased activity of mitochondrial fatty acid oxidation and oxidative phosphorylation, and decreased overall ATP production. Our findings confirm mitochondrial energy dysfunction in a patient with ITCH deficiency offering the opportunity to assess alternative therapeutic options.

19.
Cell Mol Neurobiol ; 31(5): 775-85, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21424830

RESUMEN

Methylmalonic acidemia and propionic acidemia are organic acidemias biochemically characterized by predominant tissue accumulation of methylmalonic acid (MMA) and propionic acid (PA), respectively. Affected patients present predominantly neurological symptoms, whose pathogenesis is not yet fully established. In the present study we investigated the in vitro effects of MMA and PA on important parameters of lipid and protein oxidative damage and on the production of reactive species in synaptosomes from cerebrum of developing rats. Synaptosomes correspond to nerve terminals that have been used to investigate toxic properties of compounds on neuronal cells. The in vivo effects of intrastriatal injection of MMA and PA on the same parameters and on enzymatic antioxidant defenses, were also studied. MMA-induced in vitro and in vivo lipid peroxidation and protein oxidative damage. Furthermore, the lipid oxidative damage was attenuated or prevented, pending on the doses utilized, by the free radical scavengers α-tocopherol, melatonin and by the NMDA glutamate receptor antagonist MK-801, implying the involvement of reactive species and glutamate receptor activation in these effects. In addition, 2',7'-dichlorofluorescein diacetate oxidation was significantly increased in synaptosomes by MMA, reinforcing that reactive species generation is elicited by this organic acid. We also verified that glutathione peroxidase activity was inhibited by intrastriatal MMA injection. In contrast, PA did not induce any significant effect on all parameters examined in vitro and in vivo, implying a selective action for MMA. The present data demonstrate that oxidative stress is induced by MMA in vitro in nerve terminals and in vivo in striatum, suggesting the participation of neuronal cells in MMA-elicited oxidative damage.


Asunto(s)
Antioxidantes/metabolismo , Ácido Metilmalónico/toxicidad , Neostriado/efectos de los fármacos , Neostriado/patología , Terminaciones Nerviosas/efectos de los fármacos , Terminaciones Nerviosas/patología , Estrés Oxidativo/efectos de los fármacos , Animales , Fluoresceínas/metabolismo , Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Ácido Metilmalónico/administración & dosificación , Neostriado/enzimología , Oxidación-Reducción/efectos de los fármacos , Carbonilación Proteica/efectos de los fármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
20.
Neurochem Res ; 36(2): 205-14, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20976553

RESUMEN

Lysine (Lys) accumulation in tissues and biological fluids is the biochemical hallmark of patients affected by familial hyperlysinemia (FH) and other inherited metabolic disorders. In the present study we investigated the effects of acute administration of Lys on relevant parameters of energy metabolism and oxidative stress in striatum of young rats. We verified that Lys in vivo intrastriatal injection did not change the citric acid cycle function and creatine kinase activity, but, in contrast, significantly inhibited synaptic Na(+),K(+)-ATPase activity in striatum prepared 2 and 12 h after injection. Moreover, Lys induced lipid peroxidation and diminished the concentrations of glutathione 2 h after injection. These effects were prevented by the antioxidant scavengers melatonin and the combination of α-tocopherol and ascorbic acid. Lys also inhibited glutathione peroxidase activity 12 h after injection. Therefore it is assumed that inhibition of synaptic Na(+),K(+)-ATPase and oxidative damage caused by brain Lys accumulation may possibly contribute to the neurological manifestations of FH and other neurometabolic conditions with high concentrations of this amino acid.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Lisina/farmacología , Estrés Oxidativo/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , Animales , Oxidación-Reducción , Ratas , Ratas Wistar , Sinapsis/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA