Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neurochem Res ; 49(3): 732-743, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38063948

RESUMEN

Astrocytes have key regulatory roles in central nervous system (CNS), integrating metabolic, inflammatory and synaptic responses. In this regard, type I interferon (IFN) receptor signaling in astrocytes can regulate synaptic plasticity. Simvastatin is a cholesterol-lowering drug that has shown anti-inflammatory properties, but its effects on astrocytes, a main source of cholesterol for neurons, remain to be elucidated. Herein, we investigated the effects of simvastatin in inflammatory and functional parameters of primary cortical and hypothalamic astrocyte cultures obtained from IFNα/ß receptor knockout (IFNα/ßR-/-) mice. Overall, simvastatin decreased extracellular levels of tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), which were related to a downregulation in gene expression in hypothalamic, but not in cortical astrocytes. Moreover, there was an increase in anti-inflammatory interleukin-10 (IL-10) in both structures. Effects of simvastatin in inflammatory signaling also involved a downregulation of cyclooxygenase 2 (COX-2) gene expression as well as an upregulation of nuclear factor κB subunit p65 (NFκB p65). The expression of cytoprotective genes sirtuin 1 (SIRT1) and nuclear factor erythroid derived 2 like 2 (Nrf2) was also increased by simvastatin. In addition, simvastatin increased glutamine synthetase (GS) activity and glutathione (GSH) levels only in cortical astrocytes. Our findings provide evidence that astrocytes from different regions are important cellular targets of simvastatin in the CNS, even in the absence of IFNα/ßR, which was showed by the modulation of cytokine production and release, as well as the expression of cytoprotective genes and functional parameters.


Asunto(s)
Astrocitos , Simvastatina , Ratones , Animales , Astrocitos/metabolismo , Simvastatina/farmacología , Ratones Noqueados , Factor de Necrosis Tumoral alfa/metabolismo , Interferón-alfa/metabolismo , Interferón-alfa/farmacología , Antiinflamatorios/farmacología , Colesterol/metabolismo , Células Cultivadas
2.
Neurochem Res ; 49(3): 758-770, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38104040

RESUMEN

Maple Syrup Urine Disease (MSUD) is a metabolic disease characterized by the accumulation of branched-chain amino acids (BCAA) in different tissues due to a deficit in the branched-chain alpha-ketoacid dehydrogenase complex. The most common symptoms are poor feeding, psychomotor delay, and neurological damage. However, dietary therapy is not effective. Studies have demonstrated that memantine improves neurological damage in neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. Therefore, we hypothesize that memantine, an NMDA receptor antagonist can ameliorate the effects elicited by BCAA in an MSUD animal model. For this, we organized the rats into four groups: control group (1), MSUD group (2), memantine group (3), and MSUD + memantine group (4). Animals were exposed to the MSUD model by the administration of BCAA (15.8 µL/g) (groups 2 and 4) or saline solution (0.9%) (groups 1 and 3) and treated with water or memantine (5 mg/kg) (groups 3 and 4). Our results showed that BCAA administration induced memory alterations, and changes in the levels of acetylcholine in the cerebral cortex. Furthermore, induction of oxidative damage and alterations in antioxidant enzyme activities along with an increase in pro-inflammatory cytokines were verified in the cerebral cortex. Thus, memantine treatment prevented the alterations in memory, acetylcholinesterase activity, 2',7'-Dichlorofluorescein oxidation, thiobarbituric acid reactive substances levels, sulfhydryl content, and inflammation. These findings suggest that memantine can improve the pathomechanisms observed in the MSUD model, and may improve oxidative stress, inflammation, and behavior alterations.


Asunto(s)
Enfermedad de la Orina de Jarabe de Arce , Ratas , Animales , Enfermedad de la Orina de Jarabe de Arce/tratamiento farmacológico , Enfermedad de la Orina de Jarabe de Arce/metabolismo , Memantina/farmacología , Memantina/uso terapéutico , Acetilcolinesterasa , Modelos Animales de Enfermedad , Aminoácidos de Cadena Ramificada , Antioxidantes/farmacología , Inflamación
3.
Mol Cell Biochem ; 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38498105

RESUMEN

Mucopolysaccharidosis type II (MPS II; Hunter syndrome) is a lysosomal storage disease caused by mutations in the gene encoding the enzyme iduronate 2-sulfatase (IDS) and biochemically characterized by the accumulation of glycosaminoglycans (GAGs) in different tissues. It is a multisystemic disorder that presents liver abnormalities, the pathophysiology of which is not yet established. In the present study, we evaluated bioenergetics, redox homeostasis, and mitochondrial dynamics in the liver of 6-month-old MPS II mice (IDS-). Our findings show a decrease in the activity of α-ketoglutarate dehydrogenase and an increase in the activities of succinate dehydrogenase and malate dehydrogenase. The activity of mitochondrial complex I was also increased whereas the other complex activities were not affected. In contrast, mitochondrial respiration, membrane potential, ATP production, and calcium retention capacity were not altered. Furthermore, malondialdehyde levels and 2',7'-dichlorofluorescein oxidation were increased in the liver of MPS II mice, indicating lipid peroxidation and increased ROS levels, respectively. Sulfhydryl and reduced glutathione levels, as well as glutathione S-transferase, glutathione peroxidase (GPx), superoxide dismutase, and catalase activities were also increased. Finally, the levels of proteins involved in mitochondrial mass and dynamics were decreased in knockout mice liver. Taken together, these data suggest that alterations in energy metabolism, redox homeostasis, and mitochondrial dynamics can be involved in the pathophysiology of liver abnormalities observed in MPS II.

4.
Biochem Biophys Res Commun ; 684: 149123, 2023 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-37871522

RESUMEN

Aminoacylase 1 (ACY1) deficiency is an inherited metabolic disorder biochemically characterized by high urinary concentrations of aliphatic N-acetylated amino acids and associated with a broad clinical spectrum with predominant neurological signs. Considering that the pathogenesis of ACY1 is practically unknown and the brain is highly dependent on energy production, the in vitro effects of N-acetylglutamate (NAG) and N-acetylmethionine (NAM), major metabolites accumulating in ACY1 deficiency, on the enzyme activities of the citric acid cycle (CAC), of the respiratory chain complexes and glutamate dehydrogenase (GDH), as well as on ATP synthesis were evaluated in brain mitochondrial preparations of developing rats. NAG mildly inhibited mitochondrial isocitrate dehydrogenase 2 (IDH2) activity, moderately inhibited the activities of isocitrate dehydrogenase 3 (IDH3) and complex II-III of the respiratory chain and markedly suppressed the activities of complex IV and GDH. Of note, the NAG-induced inhibitory effect on IDH3 was competitive, whereas that on GDH was mixed. On the other hand, NAM moderately inhibited the activity of respiratory complexes II-III and GDH activities and strongly decreased complex IV activity. Furthermore, NAM was unable to modify any of the CAC enzyme activities, indicating a selective effect of NAG toward IDH mitochondrial isoforms. In contrast, the activities of citrate synthase, α-ketoglutarate dehydrogenase, malate dehydrogenase, and of the respiratory chain complexes I and II were not changed by these N-acetylated amino acids. Finally, NAG and NAM strongly decreased mitochondrial ATP synthesis. Taken together, the data indicate that NAG and NAM impair mitochondrial brain energy homeostasis.


Asunto(s)
Ácido Glutámico , Isocitrato Deshidrogenasa , Ratas , Animales , Ácido Glutámico/metabolismo , Isocitrato Deshidrogenasa/metabolismo , Ratas Wistar , Metabolismo Energético , Encéfalo/metabolismo , Adenosina Trifosfato/metabolismo , Homeostasis
5.
J Neurovirol ; 29(5): 577-587, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37501054

RESUMEN

Patients affected by COVID-19 present mostly with respiratory symptoms but acute neurological symptoms are also commonly observed. Furthermore, a considerable number of individuals develop persistent and often remitting symptoms months after infection, characterizing the condition called long-COVID. Since the pathophysiology of acute and persistent neurological manifestations is not fully established, we evaluated the expression of different genes in hippocampal slices of aged rats exposed to the serum of a post-COVID (sPC) individual and to the serum of patients infected by SARS-CoV-2 [Zeta (sZeta) and Gamma (sGamma) variants]. The expression of proteins related to inflammatory process, redox homeostasis, mitochondrial quality control and glial reactivity was determined. Our data show that the exposure to sPC, sZeta and sGamma differentially altered the mRNA levels of most inflammatory proteins and reduced those of antioxidant response markers in rat hippocampus. Furthermore, a decrease in the expression of mitochondrial biogenesis genes was induced by all serum samples, whereas a reduction in mitochondrial dynamics was only caused by sPC. Regarding the glial reactivity, S100B expression was modified by sPC and sZeta. These findings demonstrate that changes in the inflammatory response and a reduction of mitochondrial biogenesis and dynamics may contribute to the neurological damage observed in COVID-19 patients.


Asunto(s)
COVID-19 , Humanos , Animales , Ratas , COVID-19/genética , Enfermedades Neuroinflamatorias , Síndrome Post Agudo de COVID-19 , SARS-CoV-2 , Homeostasis , Hipocampo
6.
Cell Mol Neurobiol ; 43(6): 2895-2907, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36862242

RESUMEN

Isolated sulfite oxidase (ISOD) and molybdenum cofactor (MoCD) deficiencies are genetic diseases biochemically characterized by the toxic accumulation of sulfite in the tissues of patients, including the brain. Neurological dysfunction and brain abnormalities are commonly observed soon after birth, and some patients also have neuropathological alterations in the prenatal period (in utero). Thus, we investigated the effects of sulfite on redox and mitochondrial homeostasis, as well as signaling proteins in the cerebral cortex of rat pups. One-day-old Wistar rats received an intracerebroventricular administration of sulfite (0.5 µmol/g) or vehicle and were euthanized 30 min after injection. Sulfite administration decreased glutathione levels and glutathione S-transferase activity, and increased heme oxygenase-1 content in vivo in the cerebral cortex. Sulfite also reduced the activities of succinate dehydrogenase, creatine kinase, and respiratory chain complexes II and II-III. Furthermore, sulfite increased the cortical content of ERK1/2 and p38. These findings suggest that redox imbalance and bioenergetic impairment induced by sulfite in the brain are pathomechanisms that may contribute to the neuropathology of newborns with ISOD and MoCD. Sulfite disturbs antioxidant defenses, bioenergetics, and signaling pathways in the cerebral cortex of neonatal rats. CII: complex II; CII-III: complex II-III; CK: creatine kinase; GST: glutathione S-transferase; HO-1: heme oxygenase-1; SDH: succinate dehydrogenase; SO32-: sulfite.


Asunto(s)
Corteza Cerebral , Metabolismo Energético , Cofactores de Molibdeno , Sulfito-Oxidasa , Sulfitos , Animales , Ratas , Animales Recién Nacidos , Oxidación-Reducción , Sulfitos/efectos adversos , Sulfito-Oxidasa/metabolismo , Cofactores de Molibdeno/metabolismo , Ratas Wistar , Homeostasis , Mitocondrias/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Antioxidantes/metabolismo
7.
Mol Cell Biochem ; 478(6): 1205-1216, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36272012

RESUMEN

Aging is intrinsically related to metabolic changes and characterized by the accumulation of oxidative and inflammatory damage, as well as alterations in gene expression and activity of several signaling pathways, which in turn impact on homeostatic responses of the body. Hypothalamus is a brain region most related to these responses, and increasing evidence has highlighted a critical role of astrocytes in hypothalamic homeostatic functions, particularly during aging process. The purpose of this study was to investigate the in vitro effects of a chronic treatment with resveratrol (1 µM during 15 days, which was replaced once every 3 days), a recognized anti-inflammatory and antioxidant molecule, in primary hypothalamic astrocyte cultures obtained from aged rats (24 months old). We observed that aging process changes metabolic, oxidative, inflammatory, and senescence parameters, as well as glial markers, while long-term resveratrol treatment prevented these effects. In addition, resveratrol upregulated key signaling pathways associated with cellular homeostasis, including adenosine receptors, nuclear factor erythroid-derived 2-like 2 (Nrf2), heme oxygenase 1 (HO-1), sirtuin 1 (SIRT1), proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), and phosphoinositide 3-kinase (PI3K). Our data corroborate the glioprotective effect of resveratrol in aged hypothalamic astrocytes, reinforcing the beneficial role of resveratrol in the aging process.


Asunto(s)
Astrocitos , Fosfatidilinositol 3-Quinasas , Ratas , Animales , Resveratrol/farmacología , Astrocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Células Cultivadas , Hipotálamo/metabolismo , Sirtuina 1/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/farmacología
8.
Metab Brain Dis ; 38(1): 287-293, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36305998

RESUMEN

Maple Syrup Urine Disease (MSUD) is an inborn error of metabolism (EIM) biochemically characterized by the tissue accumulation of branched-chain amino acids (BCAA) and their branched-chain alpha-keto acids. The mechanisms by which BCAA and their branched-chain alpha-keto acids lead to the neurological damage observed in MSUD are poorly understood. Mounting evidence has demonstrated that BCAA induce the overproduction of reactive oxygen species, which may modulate several important signaling pathways necessary for cellular homeostasis maintenance, such as autophagy. Taking this into account, we evaluated the effects of BCAA on the autophagic pathway in brain structures of rats submitted to the administration of these amino acids (animal model of MSUD). Our findings showed that BCAA significantly increased the levels of Beclin-1, ATG7, and ATG5 in the cerebral cortex of rats. In addition, BCAA augmented ATG12 levels in the striatum and ATG5 and LC3 I-II in the hippocampus. Therefore, our work demonstrates that the administration of BCAA increases autophagy and autophagic cell death, possibly mediated by the elevated levels of reactive species generated by BCAA.


Asunto(s)
Enfermedad de la Orina de Jarabe de Arce , Ratas , Animales , Enfermedad de la Orina de Jarabe de Arce/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Ratas Wistar , Modelos Animales de Enfermedad , Encéfalo/metabolismo , Cetoácidos , Autofagia
9.
Cell Mol Neurobiol ; 42(3): 565-575, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33034777

RESUMEN

Ethylmalonic encephalopathy (EE) is a severe intoxication disorder caused by mutations in the ETHE1 gene that encodes a mitochondrial sulfur dioxygenase involved in the catabolism of hydrogen sulfide. It is biochemically characterized by tissue accumulation of hydrogen sulfide and its by-product thiosulfate, as well as of ethylmalonic acid due to hydrogen sulfide-induced inhibition of short-chain acyl-CoA dehydrogenase. Patients usually present with early onset severe brain damage associated to encephalopathy, chronic hemorrhagic diarrhea and vascular lesions with petechial purpura and orthostatic acrocyanosis whose pathophysiology is poorly known. Current treatment aims to reduce hydrogen sulfide accumulation, but does not significantly prevent encephalopathy and most fatalities. In this review, we will summarize the present knowledge obtained from human and animal studies showing that disruption of mitochondrial and redox homeostasis may represent relevant pathomechanisms of tissue damage in EE. Mounting evidence show that hydrogen sulfide and ethylmalonic acid markedly disturb critical mitochondrial functions and induce oxidative stress. Novel therapeutic strategies using promising candidate drugs for this devastating disease are also discussed.


Asunto(s)
Lesiones Encefálicas , Púrpura , Animales , Encéfalo/metabolismo , Encefalopatías Metabólicas Innatas , Lesiones Encefálicas/metabolismo , Homeostasis , Humanos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Oxidación-Reducción , Púrpura/genética , Púrpura/metabolismo , Púrpura/patología
10.
Amino Acids ; 54(11): 1505-1517, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35927507

RESUMEN

Methylmalonic acidemia is a neurometabolic disorder biochemically characterized by the accumulation of methylmalonic acid (MMA) in different tissues, including the central nervous system (CNS). In this sense, it has been shown that high levels of this organic acid have a key role in the progressive neurological deterioration in patients. Astroglial cells actively participate in a wide range of CNS functions, such as antioxidant defenses and inflammatory response. Considering the role of these cells to maintain brain homeostasis, in the present study, we investigated the effects of MMA on glial parameters, focusing on redox homeostasis and inflammatory process, as well as putative mediators of these events in C6 astroglial cells. MMA decreased cell viability, glutathione levels, and antioxidant enzyme activities, increased inflammatory response, and changed the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), nuclear factor kappa B (NFκB), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), inducible nitric oxide synthase (iNOS), cyclooxygenase 2 (COX-2), and adenosine receptors, suggesting that these transcriptional factors and proteins may underlie the glial responses induced by MMA. Moreover, we also demonstrated the protective roles of melatonin and resveratrol against MMA-induced inflammation and decrease in glutathione levels. In summary, our findings support the hypothesis that astroglial changes are associated with pathogenesis of methylmalonic acidemia. In addition, we showed that these cells might be potential targets for preventive/therapeutic strategies by using molecules, such as melatonin and resveratrol, which mediated glioprotection in this inborn error of metabolism.


Asunto(s)
Melatonina , Ácido Metilmalónico , Animales , Ratas , Humanos , Resveratrol/farmacología , Astrocitos , Melatonina/farmacología , Antioxidantes/farmacología , Ratas Wistar , Oxidación-Reducción , Glutatión/farmacología , Homeostasis
11.
Nutr Neurosci ; 25(10): 2066-2076, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34076555

RESUMEN

INTRODUCTION: Polyphenols are compounds found in plants that have been extensively studied due to the health benefits of its consumption in adulthood. Meanwhile, recent evidence suggests that polyphenol consumption during pregnancy may not be safe for the fetus. OBJECTIVE: The goal of this study was to evaluate the effect of naringenin supplementation during pregnancy on brain redox homeostasis and mitochondrial activity of the newborn rat. METHODS: Adult female Wistar rats were divided into two groups: (1) vehicle (1 mL/Kg p.o.) or (2) naringenin (50 mg/Kg p.o.). Naringenin was administered once a day during pregnancy. The offspring were euthanized on postnatal day 7, as well the dams, and brain regions were dissected. RESULTS: The offspring cerebellum was the most affected region, presenting increased activity of the mitochondrial electron transport system, allied to increased reactive species levels, lipid peroxidation, and glutathione concentration. The nitric oxide levels suffered structure-dependent alteration, with decreased levels in the pups' cerebellum and increased in the hippocampus. The offspring parietal cortex was not affected, as well as the parameters evaluated in the dams' brains. CONCLUSION: Maternal consumption of naringenin alters offspring cerebellar redox homeostasis, which could be related to adverse effects on the motor and cognitive development in the descendants.


Asunto(s)
Polifenoles , Efectos Tardíos de la Exposición Prenatal , Animales , Animales Recién Nacidos , Cerebelo , Femenino , Glutatión , Homeostasis , Humanos , Óxido Nítrico , Oxidación-Reducción , Embarazo , Ratas , Ratas Wistar
12.
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
13.
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
14.
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
15.
J Inherit Metab Dis ; 44(2): 481-491, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32882059

RESUMEN

Sulfite oxidase (SO) deficiency is a disorder caused either by isolated deficiency of SO or by defects in the synthesis of its molybdenum cofactor. It is characterized biochemically by tissue sulfite accumulation. Patients present with seizures, progressive neurological damage, and basal ganglia abnormalities, the pathogenesis of which is not fully established. Treatment is supportive and largely ineffective. To address the pathophysiology of sulfite toxicity, we examined the effects of intrastriatal administration of sulfite in rats on antioxidant defenses, energy transfer, and mitogen-activated protein kinases (MAPK) and apoptosis pathways in rat striatum. Sulfite administration decreased glutathione (GSH) concentration and glutathione peroxidase, glucose-6-phosphate dehydrogenase, glutathione S-transferase, and glutathione reductase activities in striatal tissue. Creatine kinase (CK) activity, a crucial enzyme for cell energy transfer, was also decreased by sulfite. Superoxide dismutase-1 (SOD1) and catalase (CAT) proteins were increased, while heme oxygenase-1 (HO-1) was decreased. Additionally, sulfite altered phosphorylation of MAPK by decreasing of p38 and increasing of ERK. Sulfite further augmented the content of GSK-3ß, Bok, and cleaved caspase-3, indicating increased apoptosis. JP4-039 is a mitochondrial-targeted antioxidant that reaches higher intramitochondrial levels than other traditional antioxidants. Intraperitoneal injection of JP4-039 before sulfite administration preserved activity of antioxidant enzymes and CK. It also prevented or attenuated alterations in SOD1, CAT, and HO-1 protein content, as well as changes in p38, ERK, and apoptosis markers. In sum, oxidative stress and apoptosis induced by sulfite injection are prevented by JP4-039, identifying this molecule as a promising candidate for pharmacological treatment of SO-deficient patients.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/prevención & control , Antioxidantes/farmacología , Cuerpo Estriado/metabolismo , Mitocondrias/metabolismo , Óxidos de Nitrógeno/farmacocinética , Sulfito-Oxidasa/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/patología , Animales , Catalasa/metabolismo , Muerte Celular/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Creatina Quinasa/metabolismo , Transferencia de Energía/efectos de los fármacos , Glutatión Peroxidasa/metabolismo , Glutatión Peroxidasa/farmacología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Sulfitos/metabolismo , Superóxido Dismutasa/metabolismo
16.
Br J Nutr ; 123(10): 1117-1126, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32077406

RESUMEN

The study of polyphenols' effects on health has been gaining attention lately. In addition to reacting with important enzymes, altering the cell metabolism, these substances can present either positive or negative metabolic alterations depending on their consumption levels. Naringenin, a citrus flavonoid, already presents diverse metabolic effects. The objective of this work was to evaluate the effect of maternal naringenin supplementation during pregnancy on the tricarboxylic acid cycle activity in offspring's cerebellum. Adult female Wistar rats were divided into two groups: (1) vehicle (1 ml/kg by oral administration (p.o.)) or (2) naringenin (50 mg/kg p.o.). The offspring were euthanised at 7th day of life, and the cerebellum was dissected to analyse citrate synthase, isocitrate dehydrogenase (IDH), α-ketoglutarate dehydrogenase (α-KGDH) and malate dehydrogenase (MDH) activities. Molecular docking used SwissDock web server and FORECASTER Suite, and the proposed binding pose image was created on UCSF Chimera. Data were analysed by Student's t test. Naringenin supplementation during pregnancy significantly inhibited IDH, α-KGDH and MDH activities in offspring's cerebellum. A similar reduction was observed in vitro, using purified α-KGDH and MDH, subjected to pre-incubation with naringenin. Docking simulations demonstrated that naringenin possibly interacts with dehydrogenases in the substrate and cofactor binding sites, inhibiting their function. Naringenin administration during pregnancy may affect cerebellar development and must be evaluated with caution by pregnant women and their physicians.


Asunto(s)
Cerebelo/enzimología , Ciclo del Ácido Cítrico/efectos de los fármacos , Suplementos Dietéticos , Flavanonas/administración & dosificación , Fenómenos Fisiologicos Nutricionales Maternos , Animales , Citrato (si)-Sintasa/efectos de los fármacos , Femenino , Isocitrato Deshidrogenasa/efectos de los fármacos , Complejo Cetoglutarato Deshidrogenasa/efectos de los fármacos , Malato Deshidrogenasa/efectos de los fármacos , Simulación del Acoplamiento Molecular , Embarazo , Ratas , Ratas Wistar
17.
Ecotoxicol Environ Saf ; 205: 111127, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-32846293

RESUMEN

Trichlorfon is an organophosphate insecticide that is widely used on fish farms to control parasitic infections. It has been detected in freshwater ecosystems as well as in fishery products. There is a growing body of evidence to suggest that certain feed additives may reduce or prevent pesticide-induced toxicity in fish. The aim of the present study was to determine whether acute exposure to trichlorfon would alter bioenergetic homeostasis and alter fatty acid profiles in muscles of silver catfish (Rhamdia quelen). We also sought to determine whether rutin prevents or reduces these effects. Cytosolic and mitochondrial creatine kinase (CK) and activities of complexes II-III and IV in muscle were significantly inhibited by exposure to 11 mg/L trichlorfon for 48 h compared to effects in the unexposed group. Total content of polyunsaturated fatty acids (omega-3 and omega-6) were significantly lower in muscle of silver catfish exposed to 11 mg/L trichlorfon for 48 h than in the unexposed group. Addition of 3 mg rutin/kg feed increased CK activity and prevented inhibition of complex IV activity, as well as preventing all alterations of muscle fatty acid profiles elicited by exposure to trichlorfon. No significant differences were observed between groups with respect to muscle adenylate kinase or pyruvate kinase activities, as well as total content of saturated and monounsaturated fatty acids. Our findings suggest that exposure (48 h) to 11 mg trichlorfon/L water inhibits cytosolic and mitochondrial CK activity in muscle. Trichlorfon also affects activities of complexes II-III and IV in respiratory chain, with important consequences for adenosine triphosphate production. The pesticide alters fatty acid profiles in the fish and endangers human consumers of the product. The most important finding of the present study is that inclusion of rutin improves bioenergetic homeostasis and muscle fatty acid profiles, suggesting that it reduces trichlorfon-induced muscle damage.


Asunto(s)
Bagres/metabolismo , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos/metabolismo , Insecticidas/toxicidad , Músculos/efectos de los fármacos , Rutina/farmacología , Triclorfón/toxicidad , Adenosina Trifosfato/metabolismo , Adenilato Quinasa/metabolismo , Alimentación Animal , Animales , Bagres/crecimiento & desarrollo , Creatina Quinasa/metabolismo , Dieta , Aditivos Alimentarios , Homeostasis , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/metabolismo , Músculos/metabolismo
18.
Mol Biol Rep ; 46(6): 5897-5908, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31410688

RESUMEN

Phenylketonuria (PKU) is a metabolic disorder accumulating phenylalanine (Phe) and its metabolites in plasma and tissues of the patients. Regardless of the mechanisms, which Phe causes brain impairment, are poorly understood, energy deficit may have linked to the neurotoxicity in PKU. It is widely recognized that creatine is involved in maintaining of cerebral energy homeostasis. Because of this, in a previous work, we incorporated it into liposomes and this increased the concentration of creatine in the cerebral cortex. Here, we examined the effect of creatine nanoliposomes on parameters of oxidative stress, enzymes of phosphoryl transfer network, and activities of the mitochondrial respiratory chain complexes (RCC) in the cerebral cortex of young rats chemically induced hyperphenylalaninemia (HPA). HPA was induced with L-phenylalanine (5.2 µmol/g body weight; twice a day; s.c.), and phenylalanine hydroxylase inhibitor, α-methylphenylalanine (2.4 µmol/g body weight; once a day; i.p.), from the 7th to the 19th day of life. HPA reduced the activities of pyruvate kinase, creatine kinase, and complex II + III of RCC in the cerebral cortex. Creatine nanoliposomes prevented the inhibition of the activities of the complexes II + III, caused by HPA, and changes oxidative profile in the cerebral cortex. Considering the importance of the mitochondrial respiratory chain for brain energy production, our results suggesting that these nanoparticles protect against neurotoxicity caused by HPA, and can be viable candidates for treating patients HPA.


Asunto(s)
Creatina/metabolismo , Liposomas/metabolismo , Fenilcetonurias/metabolismo , Animales , Encéfalo/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Creatina/fisiología , Creatina Quinasa/metabolismo , Metabolismo Energético , Femenino , Hipocampo/metabolismo , Masculino , Nanopartículas/uso terapéutico , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Fenilalanina/metabolismo , Ratas , Ratas Wistar
19.
Microb Pathog ; 122: 53-57, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29886086

RESUMEN

Aeromonas caviae is a Gram-negative bacterium rarely found in fish but it can be associated to high mortality of infected animals. The disease pathogenesis in fish associated to liver and kidney lesions directly linked to the initiation and progression of the disease remains poorly understood. Thus, the aim of this study was to evaluate whether A. caviae infection causes oxidative stress in liver and kidney of silver catfish Rhamdia quelen, and its involvement in disease pathogenesis. Reactive oxygen species (ROS) and thiobarbituric acid reactive substances (TBARS) levels increased in liver and kidney of fish experimentally infected by A. caviae compared to the control uninfected group. On the other hand, non-protein sulfhydryl (NPSH) levels decreased in both tissues of infected animals, while the glutathione S-transferase (GST) activity decreased only in the hepatic tissue. No difference was observed between groups in both tissues regarding superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase (GR) activities and glutathione (GSH) levels. In summary, the disturbance of hepatic and renal antioxidant/oxidant equilibrium contributes to the pathophysiology of the disease in fish experimentally infected by A. caviae.


Asunto(s)
Aeromonas caviae/crecimiento & desarrollo , Antioxidantes/metabolismo , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/patología , Infecciones por Bacterias Gramnegativas/veterinaria , Oxidantes/metabolismo , Estrés Oxidativo , Animales , Bagres , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/patología , Riñón/microbiología , Riñón/patología , Hígado/microbiología , Hígado/patología , Especies Reactivas de Oxígeno/análisis
20.
Biochim Biophys Acta ; 1862(11): 2063-2074, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27523630

RESUMEN

Patients affected by sulfite oxidase (SO) deficiency present severe seizures early in infancy and progressive neurological damage, as well as tissue accumulation of sulfite, thiosulfate and S-sulfocysteine. Since the pathomechanisms involved in the neuropathology of SO deficiency are still poorly established, we evaluated the effects of sulfite on redox homeostasis and bioenergetics in cerebral cortex, striatum, cerebellum and hippocampus of rats with chemically induced SO deficiency. The deficiency was induced in 21-day-old rats by adding 200ppm of tungsten, a molybdenum competitor, in their drinking water for 9weeks. Sulfite (70mg/kg/day) was also administered through the drinking water from the third week of tungsten supplementation until the end of the treatment. Sulfite decreased reduced glutathione concentrations and the activities of glutathione reductase and glutathione S-transferase (GST) in cerebral cortex and of GST in cerebellum of SO-deficient rats. Moreover, sulfite increased the activities of complexes II and II-III in striatum and of complex II in hippocampus, but reduced the activity of complex IV in striatum of SO-deficient rats. Sulfite also decreased the mitochondrial membrane potential in cerebral cortex and striatum, whereas it had no effect on mitochondrial mass in any encephalic tissue evaluated. Finally, sulfite inhibited the activities of malate and glutamate dehydrogenase in cerebral cortex of SO-deficient rats. Taken together, our findings indicate that cerebral cortex and striatum are more vulnerable to sulfite-induced toxicity than cerebellum and hippocampus. It is presumed that these pathomechanisms may contribute to the pathophysiology of neurological damage found in patients affected by SO deficiency.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA