Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 242
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Alzheimers Dement ; 19(1): 333-342, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35522844

RESUMEN

To promote new thinking of the pathogenesis of Alzheimer's disease (AD), we examine the central role of mitochondrial dysfunction in AD. Pathologically, AD is characterized by progressive neuronal loss and biochemical abnormalities including mitochondrial dysfunction. Conventional thinking has dictated that AD is driven by amyloid beta pathology, per the Amyloid Cascade Hypothesis. However, the underlying mechanism of how amyloid beta leads to cognitive decline remains unclear. A model correctly identifying the pathogenesis of AD is critical and needed for the development of effective therapeutics. Mitochondrial dysfunction is closely linked to the core pathological feature of AD: neuronal dysfunction. Targeting mitochondria and associated proteins may hold promise for new strategies for the development of disease-modifying therapies. According to the Mitochondrial Cascade Hypothesis, mitochondrial dysfunction drives the pathogenesis of AD, as baseline mitochondrial function and mitochondrial change rates influence the progression of cognitive decline. HIGHLIGHTS: The Amyloid Cascade Model does not readily account for various parameters associated with Alzheimer's disease (AD). A unified model correctly identifying the pathogenesis of AD is greatly needed to inform the development of successful therapeutics. Mitochondria play a key and central role in the maintenance of optimal neuronal and synaptic function, the core pathological feature of AD. Mitochondrial dysfunction may be the primary cause of AD, and is a promising target for new therapeutic strategies.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Humanos , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Mitocondrias , Disfunción Cognitiva/metabolismo , Neuronas/metabolismo
2.
Neuropathol Appl Neurobiol ; 48(1): e12747, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34237158

RESUMEN

AIMS: Mitochondrial dysfunction and inflammation are at the core of axonal degeneration in several multifactorial neurodegenerative diseases, including multiple sclerosis, Alzheimer's disease, and Parkinson's disease. The transcriptional coregulator RIP140/NRIP1 (receptor-interacting protein 140) modulates these functions in liver and adipose tissue, but its role in the nervous system remains unexplored. Here, we investigated the impact of RIP140 in the Abcd1- mouse model of X-linked adrenoleukodystrophy (X-ALD), a genetic model of chronic axonopathy involving the convergence of redox imbalance, bioenergetic failure, and chronic inflammation. METHODS AND RESULTS: We provide evidence that RIP140 is modulated through a redox-dependent mechanism driven by very long-chain fatty acids (VLCFAs), the levels of which are increased in X-ALD. Genetic inactivation of RIP140 prevented mitochondrial depletion and dysfunction, bioenergetic failure, inflammatory dysregulation, axonal degeneration and associated locomotor disabilities in vivo in X-ALD mouse models. CONCLUSIONS: Together, these findings show that aberrant overactivation of RIP140 promotes neurodegeneration in X-ALD, underscoring its potential as a therapeutic target for X-ALD and other neurodegenerative disorders that present with metabolic and inflammatory dyshomeostasis.


Asunto(s)
Adrenoleucodistrofia , Miembro 1 de la Subfamilia D de Transportador de Casetes de Unión al ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/uso terapéutico , Adrenoleucodistrofia/genética , Adrenoleucodistrofia/metabolismo , Animales , Modelos Animales de Enfermedad , Homeostasis , Ratones , Mitocondrias/metabolismo , Proteína de Interacción con Receptores Nucleares 1
3.
Hum Mol Genet ; 27(16): 2874-2892, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29860433

RESUMEN

Impaired glucose metabolism, decreased levels of thiamine and its phosphate esters, and reduced activity of thiamine-dependent enzymes, such as pyruvate dehydrogenase, alpha-ketoglutarate dehydrogenase and transketolase occur in Alzheimer's disease (AD). Thiamine deficiency exacerbates amyloid beta (Aß) deposition, tau hyperphosphorylation and oxidative stress. Benfotiamine (BFT) rescued cognitive deficits and reduced Aß burden in amyloid precursor protein (APP)/PS1 mice. In this study, we examined whether BFT confers neuroprotection against tau phosphorylation and the generation of neurofibrillary tangles (NFTs) in the P301S mouse model of tauopathy. Chronic dietary treatment with BFT increased lifespan, improved behavior, reduced glycated tau, decreased NFTs and prevented death of motor neurons. BFT administration significantly ameliorated mitochondrial dysfunction and attenuated oxidative damage and inflammation. We found that BFT and its metabolites (but not thiamine) trigger the expression of Nrf2/antioxidant response element (ARE)-dependent genes in mouse brain as well as in wild-type but not Nrf2-deficient fibroblasts. Active metabolites were more potent in activating the Nrf2 target genes than the parent molecule BFT. Docking studies showed that BFT and its metabolites (but not thiamine) bind to Keap1 with high affinity. These findings demonstrate that BFT activates the Nrf2/ARE pathway and is a promising therapeutic agent for the treatment of diseases with tau pathology, such as AD, frontotemporal dementia and progressive supranuclear palsy.


Asunto(s)
Elementos de Respuesta Antioxidante/genética , Factor 2 Relacionado con NF-E2/genética , Agregación Patológica de Proteínas/tratamiento farmacológico , Tauopatías/tratamiento farmacológico , Tiamina/análogos & derivados , Péptidos beta-Amiloides/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Ratones , Ratones Transgénicos , Neuroprotección/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Transducción de Señal/efectos de los fármacos , Tauopatías/genética , Tauopatías/fisiopatología , Tiamina/administración & dosificación , Proteínas tau/genética
4.
Eur J Neurosci ; 52(4): 3242-3255, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31958881

RESUMEN

Animal models of human diseases are crucial experimental tools to investigate the mechanisms involved in disease pathogenesis and to develop new therapies. In spite of the numerous animal models currently available that reproduce several neuropathological features of Parkinson disease (PD), it is challenging to have one that consistently recapitulates human PD conditions in both motor behaviors and biochemical pathological outcomes. Given that, we have implemented a new paradigm to expose rats to a chronic low dose of paraquat (PQ), using osmotic minipumps and characterized the developed pathologic features over time. The PQ exposure paradigm used lead to a rodent model of PD depicting progressive nigrostriatal dopaminergic neurodegeneration, characterized by a 41% significant loss of dopaminergic neuron in the substantia nigra pars compacta (SNpc), a significant decrease of 18% and 40% of dopamine levels in striatum at week 5 and 8, respectively, and a significant 1.5-fold decrease in motor performance. We observed a significant increase of microglia activation state, sustained levels of α-synucleinopathy and increased oxidative stress markers in the SNpc. In summary, this is an explorative study that allowed to characterize an improved PQ-based rat model that recapitulates cardinal features of PD and may represent an attractive tool to investigate several mechanisms underlying the various aspects of PD pathogenesis as well as for the validation of the efficacy of new therapeutic approaches that targets different mechanisms involved in PD neurodegeneration.


Asunto(s)
Paraquat , Enfermedad de Parkinson , Animales , Cuerpo Estriado , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas , Paraquat/toxicidad , Porción Compacta de la Sustancia Negra , Ratas , Sustancia Negra
5.
Int J Mol Sci ; 21(23)2020 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-33255464

RESUMEN

Small biomolecules, such as coenzyme A (CoA) and acetyl coenzyme A (acetyl-CoA), play vital roles in the regulation of cellular energy metabolism. In this paper, we evaluated the delayed effect of the potent hepatotoxin thioacetamide (TAA) on the concentrations of CoA and acetyl-CoA in plasma and in different rat tissues. Administration of TAA negatively affects liver function and leads to the development of hepatic encephalopathy (HE). In our experiments, rats were administered a single intraperitoneal injection of TAA at doses of 200, 400, or 600 mg/kg. Plasma, liver, kidney, and brain samples were collected six days after the TAA administration, a period that has been suggested to allow for restoration of liver function. The concentrations of CoA and acetyl-CoA in the group of rats exposed to different doses of TAA were compared to those observed in healthy rats. The results obtained indicate that even a single administration of TAA to rats is sufficient to alter the physiological balance of CoA and acetyl-CoA in the plasma and tissues of rats for an extended period of time. The initial concentrations of CoA and acetyl-CoA were not restored even after the completion of the liver regeneration process.


Asunto(s)
Acetilcoenzima A/sangre , Coenzima A/sangre , Encefalopatía Hepática/sangre , Tioacetamida/farmacología , Acetilcoenzima A/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Coenzima A/genética , Encefalopatía Hepática/inducido químicamente , Encefalopatía Hepática/patología , Humanos , Inyecciones Intraperitoneales , Hígado/efectos de los fármacos , Hígado/patología , Regeneración Hepática/genética , Ratas , Tioacetamida/toxicidad
6.
Eur J Neurosci ; 49(4): 525-532, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30408242

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disease. Two percent of the population above the age of 60 is affected by the disease. The pathological hallmarks of PD include loss of dopaminergic neurons and the presence of Lewy bodies. Mitochondrial dysfunction and oxidative stress are thought to play a pivotal role in both sporadic and familial forms of the disease. In this review we focus on the role of mitochondrial dysfunction and oxidative stress in induced pluripotent stem cell (IPSC) models of PD.We also provide an overview of therapeutics that have been tested and some possible new therapeutics that can be tested in IPSC models of PD.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedades Mitocondriales , Modelos Neurológicos , Estrés Oxidativo , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia
7.
Neurochem Res ; 44(10): 2423-2434, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31065944

RESUMEN

In this review, we summarize the available published information on the neuroprotective effects of increasing nicotinamide adenine dinucleotide (NAD+) levels in Huntington's disease models. We discuss the rationale of potential therapeutic benefit of administering nicotinamide riboside (NR), a safe and effective NAD+ precursor. We discuss the agonistic effect on the Sirtuin1-PGC-1α-PPAR pathway as well as Sirtuin 3, which converge in improving mitochondrial function, decreasing ROS production and ameliorating bioenergetics deficits. Also, we discuss the potential synergistic effect of increasing NAD+ combined with PARPs inhibitors, as a clinical therapeutic option not only in HD, but other neurodegenerative conditions.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Sirtuinas/metabolismo , Animales , Humanos , Mitocondrias/metabolismo , NAD/metabolismo
8.
Hum Mol Genet ; 25(11): 2269-2282, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27008868

RESUMEN

Huntington's disease (HD) is a devastating illness and at present there is no disease modifying therapy or cure for it; and management of the disease is limited to a few treatment options for amelioration of symptoms. Recently, we showed that the administration of bezafibrate, a pan-PPAR agonist, increases the expression of PGC-1α and mitochondrial biogenesis, and improves phenotype and survival in R6/2 transgenic mouse model of HD. Since the R6/2 mice represent a 'truncated' huntingtin (Htt) mouse model of HD, we tested the efficacy of bezafibrate in a 'full-length' Htt mouse model, the BACHD mice. Bezafibrate treatment restored the impaired PPARγ, PPARδ, PGC-1α signaling pathway, enhanced mitochondrial biogenesis and improved antioxidant defense in the striatum of BACHD mice. Untreated BACHD mice show robust and progressive motor deficits, as well as late-onset and selective neuropathology in the striatum, which was markedly ameliorated in the BACHD mice treated with bezafibrate. Our data demonstrate the efficacy of bezafibrate in ameliorating both neuropathological features and disease phenotype in BACHD mice, and taken together with our previous studies with the R6/2 mice, highlight the strong therapeutic potential of bezafibrate for treatment of HD.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/tratamiento farmacológico , PPAR delta/biosíntesis , PPAR gamma/biosíntesis , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/biosíntesis , Animales , Bezafibrato/administración & dosificación , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Biogénesis de Organelos , PPAR delta/genética , PPAR gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/antagonistas & inhibidores , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Transducción de Señal/efectos de los fármacos
9.
Hum Mol Genet ; 25(2): 317-27, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26604138

RESUMEN

Neuroinflammation, immune reactivity and mitochondrial abnormalities are considered as causes and/or contributors to neuronal degeneration. Peroxisome proliferator-activated receptors (PPARs) regulate both inflammatory and multiple other pathways that are implicated in neurodegeneration. In the present study, we investigated the efficacy of fenofibrate (Tricor), a pan-PPAR agonist that activates PPAR-α as well as other PPARs. We administered fenofibrate to superoxide dismutase 1 (SOD1(G93A)) mice daily prior to any detectable phenotypes and then animal behavior, pathology and longevity were assessed. Treated animals showed a significant slowing of the progression of disease with weight loss attenuation, enhanced motor performance, delayed onset and survival extension. Histopathological analysis of the spinal cords showed that neuronal loss was significantly attenuated in fenofibrate-treated mice. Mitochondria were preserved as indicated by Cytochrome c immunostaining in the spinal cord, which maybe partly due to increased expression of the PPAR-γ co-activator 1-α. The total mRNA analysis revealed that neuroprotective and anti-inflammatory genes were elevated, while neuroinflammatory genes were down-regulated. This study demonstrates that the activation of PPAR-α action via fenofibrate leads to neuroprotection by both reducing neuroinflammation and protecting mitochondria, which leads to a significant increase in survival in SOD1(G93A) mice. Therefore, the development of therapeutic strategies to activate PPAR-α as well as other PPARs may lead to new therapeutic agents to slow or halt the progression of amyotrophic lateral sclerosis.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Modelos Animales de Enfermedad , Fenofibrato/farmacología , Inflamación/metabolismo , Neuronas/fisiología , PPAR alfa/agonistas , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/patología , Animales , Muerte Celular , Progresión de la Enfermedad , Femenino , Fenofibrato/inmunología , Inflamación/tratamiento farmacológico , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/inmunología , Fármacos Neuroprotectores/inmunología , Fármacos Neuroprotectores/farmacología , Médula Espinal/efectos de los fármacos , Médula Espinal/inmunología , Médula Espinal/fisiopatología
10.
J Neurosci ; 36(23): 6332-51, 2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27277809

RESUMEN

UNLABELLED: A promising approach to neurotherapeutics involves activating the nuclear-factor-E2-related factor 2 (Nrf2)/antioxidant response element signaling, which regulates expression of antioxidant, anti-inflammatory, and cytoprotective genes. Tecfidera, a putative Nrf2 activator, is an oral formulation of dimethylfumarate (DMF) used to treat multiple sclerosis. We compared the effects of DMF and its bioactive metabolite monomethylfumarate (MMF) on Nrf2 signaling and their ability to block 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced experimental Parkinson's disease (PD). We show that in vitro DMF and MMF activate the Nrf2 pathway via S-alkylation of the Nrf2 inhibitor Keap1 and by causing nuclear exit of the Nrf2 repressor Bach1. Nrf2 activation by DMF but not MMF was associated with depletion of glutathione, decreased cell viability, and inhibition of mitochondrial oxygen consumption and glycolysis rates in a dose-dependent manner, whereas MMF increased these activities in vitro However, both DMF and MMF upregulated mitochondrial biogenesis in vitro in an Nrf2-dependent manner. Despite the in vitro differences, both DMF and MMF exerted similar neuroprotective effects and blocked MPTP neurotoxicity in wild-type but not in Nrf2 null mice. Our data suggest that DMF and MMF exhibit neuroprotective effects against MPTP neurotoxicity because of their distinct Nrf2-mediated antioxidant, anti-inflammatory, and mitochondrial functional/biogenetic effects, but MMF does so without depleting glutathione and inhibiting mitochondrial and glycolytic functions. Given that oxidative damage, neuroinflammation, and mitochondrial dysfunction are all implicated in PD pathogenesis, our results provide preclinical evidence for the development of MMF rather than DMF as a novel PD therapeutic. SIGNIFICANCE STATEMENT: Almost two centuries since its first description by James Parkinson, Parkinson's disease (PD) remains an incurable disease with limited symptomatic treatment. The current study provides preclinical evidence that a Food and Drug Administration-approved drug, dimethylfumarate (DMF), and its metabolite monomethylfumarate (MMF) can block nigrostriatal dopaminergic neurodegeneration in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of PD. We elucidated mechanisms by which DMF and its active metabolite MMF activates the redox-sensitive transcription factor nuclear-factor-E2-related factor 2 (Nrf2) to upregulate antioxidant, anti-inflammatory, mitochondrial biosynthetic and cytoprotective genes to render neuroprotection via distinct S-alkylating properties and depletion of glutathione. Our data suggest that targeting Nrf2-mediated gene transcription using MMF rather than DMF is a promising approach to block oxidative stress, neuroinflammation, and mitochondrial dysfunction for therapeutic intervention in PD while minimizing side effects.


Asunto(s)
Fumaratos/uso terapéutico , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Animales , Antígenos CD/metabolismo , Línea Celular Transformada , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Fumaratos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Maleatos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Trastornos Parkinsonianos/prevención & control , Ratas , Tirosina/análogos & derivados , Tirosina/farmacología
11.
Nat Rev Neurosci ; 13(5): 293-307, 2012 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22510887

RESUMEN

Major psychiatric illnesses such as mood disorders and schizophrenia are chronic, recurrent mental illnesses that affect the lives of millions of individuals. Although these disorders have traditionally been viewed as 'neurochemical diseases', it is now clear that they are associated with impairments of synaptic plasticity and cellular resilience. Although most patients with these disorders do not have classic mitochondrial disorders, there is a growing body of evidence to suggest that impaired mitochondrial function may affect key cellular processes, thereby altering synaptic functioning and contributing to the atrophic changes that underlie the deteriorating long-term course of these illnesses. Enhancing mitochondrial function could represent an important avenue for the development of novel therapeutics and also presents an opportunity for a potentially more efficient drug-development process.


Asunto(s)
Metabolismo Energético , Trastornos Mentales/metabolismo , Trastornos Mentales/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Animales , Metabolismo Energético/fisiología , Humanos , Trastornos Mentales/terapia , Enfermedades Mitocondriales/terapia , Neuronas/metabolismo , Neuronas/patología , Sinapsis/metabolismo , Sinapsis/patología
12.
Nature ; 480(7378): 547-51, 2011 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-22056989

RESUMEN

Human pluripotent stem cells (PSCs) are a promising source of cells for applications in regenerative medicine. Directed differentiation of PSCs into specialized cells such as spinal motoneurons or midbrain dopamine (DA) neurons has been achieved. However, the effective use of PSCs for cell therapy has lagged behind. Whereas mouse PSC-derived DA neurons have shown efficacy in models of Parkinson's disease, DA neurons from human PSCs generally show poor in vivo performance. There are also considerable safety concerns for PSCs related to their potential for teratoma formation or neural overgrowth. Here we present a novel floor-plate-based strategy for the derivation of human DA neurons that efficiently engraft in vivo, suggesting that past failures were due to incomplete specification rather than a specific vulnerability of the cells. Midbrain floor-plate precursors are derived from PSCs 11 days after exposure to small molecule activators of sonic hedgehog (SHH) and canonical WNT signalling. Engraftable midbrain DA neurons are obtained by day 25 and can be maintained in vitro for several months. Extensive molecular profiling, biochemical and electrophysiological data define developmental progression and confirm identity of PSC-derived midbrain DA neurons. In vivo survival and function is demonstrated in Parkinson's disease models using three host species. Long-term engraftment in 6-hydroxy-dopamine-lesioned mice and rats demonstrates robust survival of midbrain DA neurons derived from human embryonic stem (ES) cells, complete restoration of amphetamine-induced rotation behaviour and improvements in tests of forelimb use and akinesia. Finally, scalability is demonstrated by transplantation into parkinsonian monkeys. Excellent DA neuron survival, function and lack of neural overgrowth in the three animal models indicate promise for the development of cell-based therapies in Parkinson's disease.


Asunto(s)
Trasplante de Tejido Encefálico , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/trasplante , Células Madre Embrionarias/citología , Enfermedad de Parkinson/terapia , Animales , Diferenciación Celular , Línea Celular , Supervivencia Celular , Femenino , Humanos , Macaca mulatta , Mesencéfalo/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratas , Ratas Sprague-Dawley
13.
Molecules ; 22(9)2017 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-28832533

RESUMEN

Coenzyme A (CoA) and acetyl-coenzyme A (acetyl-CoA) play essential roles in cell energy metabolism. Dysregulation of the biosynthesis and functioning of both compounds may contribute to various pathological conditions. We describe here a simple and sensitive HPLC-UV based method for simultaneous determination of CoA and acetyl-CoA in a variety of biological samples, including cells in culture, mouse cortex, and rat plasma, liver, kidney, and brain tissues. The limits of detection for CoA and acetyl-CoA are >10-fold lower than those obtained by previously described HPLC procedures, with coefficients of variation <1% for standard solutions, and 1-3% for deproteinized biological samples. Recovery is 95-97% for liver extracts spiked with Co-A and acetyl-CoA. Many factors may influence the tissue concentrations of CoA and acetyl-CoA (e.g., age, fed, or fasted state). Nevertheless, the values obtained by the present HPLC method for the concentration of CoA and acetyl-CoA in selected rodent tissues are in reasonable agreement with literature values. The concentrations of CoA and acetyl-CoA were found to be very low in rat plasma, but easily measurable by the present HPLC method. The method should be useful for studying cellular energy metabolism under normal and pathological conditions, and during targeted drug therapy treatment.


Asunto(s)
Acetilcoenzima A/sangre , Acetilcoenzima A/química , Cromatografía Líquida de Alta Presión , Coenzima A/sangre , Coenzima A/química , Espectrofotometría Ultravioleta , Animales , Línea Celular , Corteza Cerebral/enzimología , Femenino , Humanos , Ratones , Ratas
14.
J Neurochem ; 139 Suppl 1: 216-231, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27546335

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disease. About 2% of the population above the age of 60 is affected by the disease. The pathological hallmarks of the disease include the loss of dopaminergic neurons in the substantia nigra and the presence of Lewy bodies that are made of α-synuclein. Several theories have been suggested for the pathogenesis of PD, of which mitochondrial dysfunction plays a pivotal role in both sporadic and familial forms of the disease. Dysfunction of the mitochondria that is caused by bioenergetic defects, mutations in mitochondrial DNA, nuclear DNA gene mutations linked to mitochondria, and changes in dynamics of the mitochondria such fusion or fission, changes in size and morphology, alterations in trafficking or transport, altered movement of mitochondria, impairment of transcription, and the presence of mutated proteins associated with mitochondria are implicated in PD. In this review, we provide a detailed overview of the mechanisms that can cause mitochondrial dysfunction in PD. We bring to the forefront, new signaling pathways such as the retromer-trafficking pathway and its implication in the disease and also provide a brief overview of therapeutic strategies to improve mitochondrial defects in PD. Bioenergetic defects, mutations in mitochondrial DNA, nuclear DNA gene mutations, alterations in mitochondrial dynamics, alterations in trafficking/transport and mitochondrial movement, abnormal size and morphology, impairment of transcription and the presence of mutated proteins associated with mitochondria are implicated in PD. In this review, we focus on the mechanisms underlying mitochondrial dysfunction in PD and bring to the forefront new signaling pathways that may be involved in PD. We also provide an overview of therapeutic strategies to improve mitochondrial defects in PD. This article is part of a special issue on Parkinson disease.


Asunto(s)
Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Animales , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Humanos , Dinámicas Mitocondriales/fisiología , Mutación/genética , Estrés Oxidativo/fisiología
15.
J Neurochem ; 139(5): 823-838, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27580471

RESUMEN

Reductions in metabolism and excess oxidative stress are prevalent in multiple neurodegenerative diseases. The activity of the mitochondrial enzyme α-ketoglutarate dehydrogenase complex (KGDHC) appears central to these abnormalities. KGDHC is diminished in multiple neurodegenerative diseases. KGDHC can not only be rate limiting for NADH production and for substrate level phosphorylation, but is also a source of reactive oxygen species (ROS). The goal of these studies was to determine how changes in KGDHC modify baseline ROS, the ability to buffer ROS, baseline glutathionylation, calcium modulation and cell death in response to external oxidants. In vivo, reducing KGDHC with adeno virus diminished neurogenesis and increased oxidative stress. In vitro, treatments of short duration increased ROS and glutathionylation and enhanced the ability of the cells to diminish the ROS from added oxidants. However, long-term reductions lessened the ability to diminish ROS, diminished glutathionylation and exaggerated oxidant-induced changes in calcium and cell death. Increasing KGDHC enhanced the ability of the cells to diminish externally added ROS and protected against oxidant-induced changes in calcium and cell death. The results suggest that brief periods of diminished KGDHC are protective, while prolonged reductions are harmful. Furthermore, elevated KGDHC activities are protective. Thus, mitogenic therapies that increase KGDHC may be beneficial in neurodegenerative diseases. Read the Editorial Highlight for this article on Page 689.


Asunto(s)
Complejo Cetoglutarato Deshidrogenasa/deficiencia , Enfermedades Neurodegenerativas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Hipocampo/metabolismo , Hipocampo/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedades Neurodegenerativas/patología
16.
Hum Mol Genet ; 23(14): 3716-32, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24556215

RESUMEN

Methylene blue (MB, methylthioninium chloride) is a phenothiazine that crosses the blood brain barrier and acts as a redox cycler. Among its beneficial properties are its abilities to act as an antioxidant, to reduce tau protein aggregation and to improve energy metabolism. These actions are of particular interest for the treatment of neurodegenerative diseases with tau protein aggregates known as tauopathies. The present study examined the effects of MB in the P301S mouse model of tauopathy. Both 4 mg/kg MB (low dose) and 40 mg/kg MB (high dose) were administered in the diet ad libitum from 1 to 10 months of age. We assessed behavior, tau pathology, oxidative damage, inflammation and numbers of mitochondria. MB improved the behavioral abnormalities and reduced tau pathology, inflammation and oxidative damage in the P301S mice. These beneficial effects were associated with increased expression of genes regulated by NF-E2-related factor 2 (Nrf2)/antioxidant response element (ARE), which play an important role in antioxidant defenses, preventing protein aggregation, and reducing inflammation. The activation of Nrf2/ARE genes is neuroprotective in other transgenic mouse models of neurodegenerative diseases and it appears to be an important mediator of the neuroprotective effects of MB in P301S mice. Moreover, we used Nrf2 knock out fibroblasts to show that the upregulation of Nrf2/ARE genes by MB is Nrf2 dependent and not due to secondary effects of the compound. These findings provide further evidence that MB has important neuroprotective effects that may be beneficial in the treatment of human neurodegenerative diseases with tau pathology.


Asunto(s)
Azul de Metileno/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Tauopatías/tratamiento farmacológico , Proteínas tau/genética , Proteínas tau/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Línea Celular , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Azul de Metileno/administración & dosificación , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Factores Sexuales , Transducción de Señal/efectos de los fármacos , Tauopatías/patología
17.
Anal Biochem ; 494: 52-4, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26576832

RESUMEN

α-Ketoglutaramate is an important glutamine metabolite in mammals, plants, and many bacteria. It is also a nicotine metabolite in certain bacteria. Previously published methods for the determination of α-ketoglutaramate in biological samples have considerable drawbacks. Here, we describe a relatively simple high-performance liquid chromatography (HPLC)-based method for measurement of α-ketoglutaramate in plasma and deproteinized tissues that overcomes these drawbacks. Concentrations of α-ketoglutaramate in normal rat liver, kidney, brain, and plasma were found to be approximately 216, 13, 6, and 19 µM, respectively. The HPLC method should be useful for studying the role of α-ketoglutaramate in eukaryotic glutamine metabolism and in bacterial nicotine metabolism.


Asunto(s)
Técnicas de Química Analítica/métodos , Cromatografía Líquida de Alta Presión , Ácidos Cetoglutáricos/análisis , Animales , Encéfalo/metabolismo , Ácidos Cetoglutáricos/sangre , Riñón/metabolismo , Hígado/metabolismo , Ratas
18.
Anal Biochem ; 503: 8-10, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27001310

RESUMEN

Here we describe a simple high-performance liquid chromatography (HPLC) procedure for the simultaneous detection and quantitation in standard solutions of 13 important metabolites of cellular energy metabolism, including 9 tricarboxylic acid (TCA) cycle components and 4 additional metabolites. The metabolites are detected by their absorbance at 210 nm. The procedure does not require prior derivatization, and an analysis can be carried out at ambient temperature within 15 min. The significance of the current work is that the current HPLC procedure should motivate the development of simplified TCA cycle enzyme assays, isotopomer analysis, and determination of selected TCA metabolite levels in plasma/tissues.


Asunto(s)
Ácidos Carboxílicos/sangre , Cromatografía Líquida de Alta Presión/métodos , Ciclo del Ácido Cítrico , Rayos Ultravioleta , Animales , Ácidos Carboxílicos/metabolismo , Femenino , Ratas , Ratas Wistar , Espectrofotometría Ultravioleta
19.
PLoS Genet ; 9(10): e1003845, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098148

RESUMEN

Macroautophagy is a conserved mechanism for the bulk degradation of proteins and organelles. Pathological studies have implicated defective macroautophagy in neurodegeneration, but physiological functions of macroautophagy in adult neurons remain unclear. Here we show that Atg7, an essential macroautophagy component, regulates dopaminergic axon terminal morphology. Mature Atg7-deficient midbrain dopamine (DA) neurons harbored selectively enlarged axonal terminals. This contrasted with the phenotype of DA neurons deficient in Pten - a key negative regulator of the mTOR kinase signaling pathway and neuron size - that displayed enlarged soma but unaltered axon terminals. Surprisingly, concomitant deficiency of both Atg7 and Pten led to a dramatic enhancement of axon terminal enlargement relative to Atg7 deletion alone. Similar genetic interactions between Atg7 and Pten were observed in the context of DA turnover and DA-dependent locomotor behaviors. These data suggest a model for morphological regulation of mature dopaminergic axon terminals whereby the impact of mTOR pathway is suppressed by macroautophagy.


Asunto(s)
Proteínas Asociadas a Microtúbulos/genética , Fosfohidrolasa PTEN/genética , Terminales Presinápticos/metabolismo , Serina-Treonina Quinasas TOR/genética , Animales , Autofagia/genética , Proteína 7 Relacionada con la Autofagia , Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/metabolismo , Morfogénesis/genética , Actividad Motora/genética , Actividad Motora/fisiología , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo
20.
Proc Natl Acad Sci U S A ; 110(12): E1102-11, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23487751

RESUMEN

Oxidative stress is a widely recognized cause of cell death associated with neurodegeneration, inflammation, and aging. Tyrosine nitration in these conditions has been reported extensively, but whether tyrosine nitration is a marker or plays a role in the cell-death processes was unknown. Here, we show that nitration of a single tyrosine residue on a small proportion of 90-kDa heat-shock protein (Hsp90), is sufficient to induce motor neuron death by the P2X7 receptor-dependent activation of the Fas pathway. Nitrotyrosine at position 33 or 56 stimulates a toxic gain of function that turns Hsp90 into a toxic protein. Using an antibody that recognizes the nitrated Hsp90, we found immunoreactivity in motor neurons of patients with amyotrophic lateral sclerosis, in an animal model of amyotrophic lateral sclerosis, and after experimental spinal cord injury. Our findings reveal that cell death can be triggered by nitration of a single protein and highlight nitrated Hsp90 as a potential target for the development of effective therapies for a large number of pathologies.


Asunto(s)
Muerte Celular/fisiología , Proteínas HSP90 de Choque Térmico/metabolismo , Ácido Peroxinitroso/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Ratas , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Tirosina/metabolismo , Receptor fas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA