Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 26(24): 4929-4936, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29040550

RESUMO

Inherited mitochondrial optic neuropathies, such as Leber's hereditary optic neuropathy (LHON) and Autosomal dominant optic atrophy (ADOA) are caused by mutant mitochondrial proteins that lead to defects in mitochondrial complex 1-driven ATP synthesis, and cause specific retinal ganglion cell (RGC) loss. Complex 1 defects also occur in patients with primary open angle glaucoma (POAG), in which there is specific RGC loss. The treatment of mitochondrial optic neuropathy in the US is only supportive. The Ndufs4 knockout (Ndufs4 KO) mouse is a mitochondrial complex 1-deficient model that leads to RGC loss and rapid vision loss and allows for streamlined testing of potential therapeutics. Preceding RGC loss in the Ndufs4 KO is the loss of starburst amacrine cells, which may be an important target in the mechanism of complex 1-deficient vision loss. Papaverine and zolpidem were recently shown to be protective of bioenergetic loss in cell models of optic neuropathy. Treatment of Ndufs4 KO mice with papaverine, zolpidem, and rapamycin-suppressed inflammation, prevented cell death, and protected from vision loss. Thus, in the Ndufs4 KO mouse model of mitochondrial optic neuropathy, papaverine and zolpidem provided significant protection from multiple pathophysiological features, and as approved drugs in wide human use could be considered for the novel indication of human optic neuropathy.


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Doenças do Nervo Óptico/metabolismo , Animais , DNA Mitocondrial/metabolismo , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/deficiência , Complexo I de Transporte de Elétrons/genética , Glaucoma de Ângulo Aberto/metabolismo , Humanos , Inflamação/metabolismo , Complexo Mediador/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Doenças Mitocondriais/metabolismo , Doenças do Nervo Óptico/genética , Papaverina/farmacologia , Piridinas/farmacologia , Células Ganglionares da Retina/metabolismo , Zolpidem
2.
Hum Mol Genet ; 26(14): 2627-2633, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28444186

RESUMO

Friedreich's ataxia (FRDA) is a neurodegenerative disease caused by inherited deficiency of the mitochondrial protein Frataxin (FXN), which has no approved therapy and is an area in which biomarkers are needed for clinical development. Here, we investigated the consequences of FXN deficiency in patient-derived FRDA fibroblast cell models, the FRDA mouse model KIKO, and in whole blood collected from patients with FRDA. We observed decreased mitochondrial copy number in all the three FRDA models tested: cells, mice and patient blood. In addition, we observed 40% residual mitochondrial gene expression in FRDA patient blood. These deficiencies of mitochondrial biogenesis in FRDA cells and patient blood are significantly correlated with FXN expression, consistent with the idea that the decreased mitochondrial biogenesis is a consequence of FXN deficiency. The observations appear relevant to the FRDA pathophysiological mechanism, as FXN-dependent deficiency in mitochondrial biogenesis and consequent mitochondrial bioenergetic defect could contribute to the neurodegenerative process. The observations may also have translational potential, as mitochondrial biogenesis could now be followed as a clinical biomarker of FRDA as a correlate of disease severity, progression, and therapeutic effect. Also, mitochondrial copy number in blood is objective, scalar and more investigator-independent than clinical-neurological patient rating scales. Thus, FXN deficiency causes mitochondrial deficiency in FRDA cells, the KIKO mouse model, and in whole blood of patients with FRDA, and this deficiency could potentially be used in clinical trial design.


Assuntos
Proteínas de Ligação ao Ferro/metabolismo , Mitocôndrias/metabolismo , Biogênese de Organelas , Animais , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Ataxia de Friedreich/genética , Expressão Gênica , Genes Mitocondriais , Humanos , Proteínas de Ligação ao Ferro/genética , Camundongos , Mitocôndrias/genética , Proteínas Mitocondriais/metabolismo , Frataxina
3.
Hum Mol Genet ; 24(10): 2848-60, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25652399

RESUMO

Mitochondrial complex I (NADH dehydrogenase) is a major contributor to neuronal energetics, and mutations in complex I lead to vision loss. Functional, neuroanatomical and transcriptional consequences of complex I deficiency were investigated in retinas of the Ndufs4 knockout mouse. Whole-eye ERGs and multielectrode arrays confirmed a major retinal ganglion cell functional loss at P32, and retinal ganglion cell loss at P42. RNAseq demonstrated a mild and then sharp increase in innate immune and inflammatory retinal transcripts at P22 and P33, respectively, which were confirmed with QRT-PCR. Intraperitoneal injection of the inflammogen lipopolysaccharide further reduced retinal ganglion cell function in Ndufs4 KO, supporting the connection between inflammatory activation and functional loss. Complex I deficiency in the retina clearly caused innate immune and inflammatory markers to increase coincident with loss of vision, and RGC functional loss. How complex I incites inflammation and functional loss is not clear, but could be the result of misfolded complex I generating a 'non-self' response, and induction of innate immune response transcripts was observed before functional loss at P22, including ß-2 microglobulin and Cx3cr1, and during vision loss at P31 (B2m, Tlr 2, 3, 4, C1qa, Cx3cr1 and Fas). These data support the hypothesis that mitochondrial complex I dysfunction in the retina triggers an innate immune and inflammatory response that results in loss of retinal ganglion cell function and death, as in Leber's hereditary Optic Neuropathy and suggests novel therapeutic routes to counter mitochondrial defects that contribute to vision loss.


Assuntos
Complexo I de Transporte de Elétrons/deficiência , Doenças Mitocondriais/fisiopatologia , Retina/fisiopatologia , Células Ganglionares da Retina/fisiologia , Animais , Morte Celular , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/imunologia , Feminino , Técnicas de Inativação de Genes , Imunidade Inata/genética , Inflamação/genética , Masculino , Camundongos , Camundongos Knockout , Doenças Mitocondriais/genética , Doenças Mitocondriais/imunologia , Retina/imunologia
4.
J Neurosci ; 35(6): 2516-29, 2015 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-25673846

RESUMO

Rett syndrome (RTT) is an autism spectrum disorder caused by loss-of-function mutations in the gene encoding MeCP2, an epigenetic modulator that binds the methyl CpG dinucleotide in target genes to regulate transcription. Previously, we and others reported a role of microglia in the pathophysiology of RTT. To understand the mechanism of microglia dysfunction in RTT, we identified a MeCP2 target gene, SLC38A1, which encodes a major glutamine transporter (SNAT1), and characterized its role in microglia. We found that MeCP2 acts as a microglia-specific transcriptional repressor of SNAT1. Because glutamine is mainly metabolized in the mitochondria, where it is used as an energy substrate and a precursor for glutamate production, we hypothesize that SNAT1 overexpression in MeCP2-deficient microglia would impair the glutamine homeostasis, resulting in mitochondrial dysfunction as well as microglial neurotoxicity because of glutamate overproduction. Supporting this hypothesis, we found that MeCP2 downregulation or SNAT1 overexpression in microglia resulted in (1) glutamine-dependent decrease in microglial viability, which was corroborated by reduced microglia counts in the brains of MECP2 knock-out mice; (2) proliferation of mitochondria and enhanced mitochondrial production of reactive oxygen species; (3) increased oxygen consumption but decreased ATP production (an energy-wasting state); and (4) overproduction of glutamate that caused NMDA receptor-dependent neurotoxicity. The abnormalities could be rectified by mitochondria-targeted expression of catalase and a mitochondria-targeted peptide antioxidant, Szeto-Schiller 31. Our results reveal a novel mechanism via which MeCP2 regulates bioenergetic pathways in microglia and suggest a therapeutic potential of mitochondria-targeted antioxidants for RTT.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Microglia/metabolismo , Doenças Mitocondriais/metabolismo , Síndromes Neurotóxicas/metabolismo , Síndrome de Rett/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Ácido Glutâmico/metabolismo , Glicina/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Consumo de Oxigênio/fisiologia , Cultura Primária de Células
5.
Hum Mol Genet ; 23(25): 6848-62, 2014 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-25113747

RESUMO

Inherited deficiency in the mitochondrial protein frataxin (FXN) causes the rare disease Friedreich's ataxia (FA), for which there is no successful treatment. We identified a redox deficiency in FA cells and used this to model the disease. We screened a 1600-compound library to identify existing drugs, which could be of therapeutic benefit. We identified the topical anesthetic dyclonine as protective. Dyclonine increased FXN transcript and FXN protein dose-dependently in FA cells and brains of animal models. Dyclonine also rescued FXN-dependent enzyme deficiencies in the iron-sulfur enzymes, aconitase and succinate dehydrogenase. Dyclonine induces the Nrf2 [nuclear factor (erythroid-derived 2)-like 2] transcription factor, which we show binds an upstream response element in the FXN locus. Additionally, dyclonine also inhibited the activity of histone methyltransferase G9a, known to methylate histone H3K9 to silence FA chromatin. Chronic dosing in a FA mouse model prevented a performance decline in balance beam studies. A human clinical proof-of-concept study was completed in eight FA patients dosed twice daily using a 1% dyclonine rinse for 1 week. Six of the eight patients showed an increase in buccal cell FXN levels, and fold induction was significantly correlated with disease severity. Dyclonine represents a novel therapeutic strategy that can potentially be repurposed for the treatment of FA.


Assuntos
Anestésicos Locais/farmacologia , Ataxia de Friedreich/tratamento farmacológico , Proteínas de Ligação ao Ferro/agonistas , Mucosa Bucal/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/agonistas , Fármacos Neuroprotetores/farmacologia , Propiofenonas/farmacologia , Aconitato Hidratase/genética , Aconitato Hidratase/metabolismo , Animais , Linhagem Celular , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Cerebelo/patologia , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Ataxia de Friedreich/patologia , Regulação da Expressão Gênica , Ensaios de Triagem em Larga Escala , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Proteínas de Ligação ao Ferro/genética , Proteínas de Ligação ao Ferro/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mucosa Bucal/metabolismo , Mucosa Bucal/patologia , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Equilíbrio Postural/efeitos dos fármacos , Transdução de Sinais , Bibliotecas de Moléculas Pequenas/farmacologia , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo , Frataxina
6.
Biol Reprod ; 95(2): 34, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27335066

RESUMO

Stallion sperm rely primarily on oxidative phosphorylation for production of ATP used in sperm motility and metabolism. The objective of the study was to identify which substrates included in Biggers, Whitten, and Whittingham (BWW) media are key to optimal mitochondrial function through measurements of sperm motility parameters, mitochondrial oxygen consumption, and cellular reactive oxygen species (ROS) production. It was expected that mitochondrial substrates, pyruvate and lactate, would support sperm motility and mitochondrial function better than the glycolytic substrate, glucose, due to direct utilization within the mitochondria. Measurements were performed after incubation in modified BWW media with varying concentrations of lactate, pyruvate, and glucose. The effects of media and duration of incubation on sperm motility, ROS production, and oxygen consumption were determined using a linear mixed-effects model. Duplicate ejaculates from four stallions were used in three separate experiments to determine the effects of substrate availability and concentration on sperm motility and mitochondrial function and the relationship of oxygen consumption with cellular ROS production. The present results indicate that lactate and pyruvate are the most important sources of energy for stallion sperm motility and velocity, and elicit a dose-dependent response. Additionally, lactate and pyruvate are ideal for maximal mitochondrial function, as sperm in these media operate at a very high level of their bioenergetic capability due to the high rate of energy metabolism. Moreover, we found that addition of glucose to the media is not necessary for short-term storage of equine sperm, and may even result in reduction of mitochondrial function. Finally, we have confirmed that ROS production can be the result of mitochondrial dysfunction as well as intense mitochondrial activity.


Assuntos
Ácido Láctico/farmacologia , Mitocôndrias/efeitos dos fármacos , Ácido Pirúvico/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Motilidade dos Espermatozoides/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Metabolismo Energético/efeitos dos fármacos , Glucose/farmacologia , Cavalos , Masculino , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Espermatozoides/metabolismo
7.
Hum Mol Genet ; 21(23): 5147-58, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22949510

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder in the developed world, and is characterized by the loss of dopaminergic (DA) neurons in the substantia nigra (SN). Somatic mitochondrial DNA (mtDNA) deletions reach their highest concentration with age in the SN in humans, and may contribute to PD; yet whether mtDNA deletions cause DA neuron degeneration remains unclear. Inherited mutations of Twinkle helicase involved in mtDNA replication causes a dominant increase in mtDNA deletions in humans. We constructed a mouse model expressing mutant Twinkle in DA neurons. Mutant mice had an increase in age-related mtDNA deletions, reduction of DA neuron number in SN at 17-22 months and displayed abnormalities in rota-rod behavior. Functional analysis of midbrain indicated a slight reduction in mitochondrial state II respiration in mutants, but no decrease in maximal respiration. Also, Parkin expression was significantly decreased in DA neurons in the SN of 22-month-old mutant mice, and in PC12 cells after 48 h transfection of mutant Twinkle. Both confocal imaging and coimmunoprecipitation indicated interaction of Twinkle with Parkin in the mitochondria. Parkin overexpression rescued the reduction of proteasome activity caused by mutant Twinkle in PC12 cells. In addition, the autophagy marker LC3 was increased in the SN of 22-month transgenics, and this increase was similarly mutant Twinkle-dependent in PC12 cells. Collectively, our data demonstrate that mammalian Twinkle is important for mitochondrial integrity in DA neurons and provide a novel mouse model in which increased mtDNA deletions may lead to DA neuron degeneration and parkinsonism.


Assuntos
DNA Helicases/genética , DNA Mitocondrial/genética , Neurônios Dopaminérgicos/metabolismo , Proteínas Mitocondriais/genética , Mutação , Doença de Parkinson/genética , Animais , Autofagia/genética , Comportamento Animal , Linhagem Celular , Respiração Celular/genética , DNA Helicases/metabolismo , Neurônios Dopaminérgicos/patologia , Expressão Gênica , Ordem dos Genes , Marcação de Genes , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Mitocondriais/metabolismo , Doença de Parkinson/metabolismo , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Substância Negra/metabolismo , Substância Negra/patologia , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
8.
Biochem Pharmacol ; 219: 115953, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38036191

RESUMO

The pharmacological interest in mitochondria is very relevant since these crucial organelles are involved in the pathogenesis of multiple diseases, such as cancer. In order to modulate cellular redox/oxidative balance and enhance mitochondrial function, numerous polyphenolic derivatives targeting mitochondria have been developed. Still, due to the drug resistance emergence in several cancer therapies, significant efforts are being made to develop drugs that combine the induction of mitochondrial metabolic reprogramming with the ability to generate reactive oxygen species, taking into consideration the varying metabolic profiles of different cell types. We previously developed a mitochondria-targeted antioxidant (AntiOxCIN6) by linking caffeic acid to lipophilic triphenylphosphonium cation through a 10-carbon aliphatic chain. The antioxidant activity of AntiOxCIN6 has been documented but how the mitochondriotropic compound impact energy metabolism of both normal and cancer cells remains unknown. We demonstrated that AntiOxCIN6 increased antioxidant defense system in HepG2 cells, although ROS clearance was ineffective. Consequently, AntiOxCIN6 significantly decreased mitochondrial function and morphology, culminating in a decreased capacity in complex I-driven ATP production without affecting cell viability. These alterations were accompanied by an increase in glycolytic fluxes. Additionally, we demonstrate that AntiOxCIN6 sensitized A549 adenocarcinoma cells for CIS-induced apoptotic cell death, while AntiOxCIN6 appears to cause metabolic changes or a redox pre-conditioning on lung MRC-5 fibroblasts, conferring protection against cisplatin. We propose that length and hydrophobicity of the C10-TPP+ alkyl linker play a significant role in inducing mitochondrial and cellular toxicity, while the presence of the antioxidant caffeic acid appears to be responsible for activating cytoprotective pathways.


Assuntos
Antioxidantes , Doenças Mitocondriais , Humanos , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Cisplatino/farmacologia , Metabolismo Energético , Espécies Reativas de Oxigênio/metabolismo , Doenças Mitocondriais/metabolismo , Pulmão/metabolismo
9.
Commun Biol ; 7(1): 195, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38366025

RESUMO

The Ketogenic Diet (KD) improves memory and longevity in aged C57BL/6 mice. We tested 7 months KD vs. control diet (CD) in the mouse Alzheimer's Disease (AD) model APP/PS1. KD significantly rescued Long-Term-Potentiation (LTP) to wild-type levels, not by changing Amyloid-ß (Aß) levels. KD's 'main actor' is thought to be Beta-Hydroxy-butyrate (BHB) whose levels rose significantly in KD vs. CD mice, and BHB itself significantly rescued LTP in APP/PS1 hippocampi. KD's 6 most significant pathways induced in brains by RNAseq all related to Synaptic Plasticity. KD induced significant increases in synaptic plasticity enzymes p-ERK and p-CREB in both sexes, and of brain-derived neurotrophic factor (BDNF) in APP/PS1 females. We suggest KD rescues LTP through BHB's enhancement of synaptic plasticity. LTP falls in Mild-Cognitive Impairment (MCI) of human AD. KD and BHB, because they are an approved diet and supplement respectively, may be most therapeutically and translationally relevant to the MCI phase of Alzheimer's Disease.


Assuntos
Doença de Alzheimer , Dieta Cetogênica , Humanos , Camundongos , Animais , Idoso , Potenciação de Longa Duração , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Camundongos Transgênicos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal
10.
Geroscience ; 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38753230

RESUMO

Genetically heterogeneous UM-HET3 mice born in 2020 were used to test possible lifespan effects of alpha-ketoglutarate (AKG), 2,4-dinitrophenol (DNP), hydralazine (HYD), nebivolol (NEBI), 16α-hydroxyestriol (OH_Est), and sodium thiosulfate (THIO), and to evaluate the effects of canagliflozin (Cana) when started at 16 months of age. OH_Est produced a 15% increase (p = 0.0001) in median lifespan in males but led to a significant (7%) decline in female lifespan. Cana, started at 16 months, also led to a significant increase (14%, p = 0.004) in males and a significant decline (6%, p = 0.03) in females. Cana given to mice at 6 months led, as in our previous study, to an increase in male lifespan without any change in female lifespan, suggesting that this agent may lead to female-specific late-life harm. We found that blood levels of Cana were approximately 20-fold higher in aged females than in young males, suggesting a possible mechanism for the sex-specific disparities in its effects. NEBI was also found to produce a female-specific decline (4%, p = 0.03) in lifespan. None of the other tested drugs provided a lifespan benefit in either sex. These data bring to 7 the list of ITP-tested drugs that induce at least a 10% lifespan increase in one or both sexes, add a fourth drug with demonstrated mid-life benefits on lifespan, and provide a testable hypothesis that might explain the sexual dimorphism in lifespan effects of the SGLT2 inhibitor Cana.

11.
Bioorg Med Chem ; 21(8): 2346-2354, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23473946

RESUMO

The effect of the alkyl side chain length of coenzyme Q10 on mitochondrial respiratory chain function has been investigated by the use of synthetic ubiquinone derivatives. Three analogues (3, 4 and 6) were identified that exhibited significantly improved effects on mitochondrial oxygen consumption and mitochondrial membrane potential, and also conferred significant cytoprotection on cultured mammalian cells in which glutathione had been depleted by treatment with diethyl maleate. The analogues also exhibited lesser inhibition of the electron transport chain than idebenone. The results obtained provide guidance for the design of CoQ10 analogues with improved activity compared to that of idebenone (1), the latter of which is undergoing evaluation in the clinic as a therapeutic agent.


Assuntos
Transporte de Elétrons/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Ubiquinona/análogos & derivados , Animais , Bovinos , Linhagem Celular , Linhagem Celular Tumoral , Citoproteção , Transporte de Elétrons/fisiologia , Humanos , Leucemia/metabolismo , Leucemia/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Relação Estrutura-Atividade , Ubiquinona/química , Ubiquinona/metabolismo , Ubiquinona/farmacologia
12.
J Biol Chem ; 285(1): 142-52, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19861415

RESUMO

Bioactive compounds reported to stimulate mitochondrial biogenesis are linked to many health benefits such increased longevity, improved energy utilization, and protection from reactive oxygen species. Previously studies have shown that mice and rats fed diets lacking in pyrroloquinoline quinone (PQQ) have reduced mitochondrial content. Therefore, we hypothesized that PQQ can induce mitochondrial biogenesis in mouse hepatocytes. Exposure of mouse Hepa1-6 cells to 10-30 microm PQQ for 24-48 h resulted in increased citrate synthase and cytochrome c oxidase activity, Mitotracker staining, mitochondrial DNA content, and cellular oxygen respiration. The induction of this process occurred through the activation of cAMP response element-binding protein (CREB) and peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha), a pathway known to regulate mitochondrial biogenesis. PQQ exposure stimulated phosphorylation of CREB at serine 133, activated the promoter of PGC-1alpha, and increased PGC-1alpha mRNA and protein expression. PQQ did not stimulate mitochondrial biogenesis after small interfering RNA-mediated reduction in either PGC-1alpha or CREB expression. Consistent with activation of the PGC-1alpha pathway, PQQ increased nuclear respiratory factor activation (NRF-1 and NRF-2) and Tfam, TFB1M, and TFB2M mRNA expression. Moreover, PQQ protected cells from mitochondrial inhibition by rotenone, 3-nitropropionic acid, antimycin A, and sodium azide. The ability of PQQ to stimulate mitochondrial biogenesis accounts in part for action of this compound and suggests that PQQ may be beneficial in diseases associated with mitochondrial dysfunction.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Cofator PQQ/farmacologia , Transativadores/genética , Animais , Bovinos , Respiração Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Camundongos , Mitocôndrias/enzimologia , Fatores Nucleares Respiratórios/metabolismo , Oxirredução/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Succinato Desidrogenase/biossíntese , Superóxidos/metabolismo , Transativadores/metabolismo , Fatores de Transcrição
13.
J Biol Chem ; 285(2): 1153-65, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19892704

RESUMO

A decrease in reactive oxygen species (ROS) production has been associated with extended life span in animal models of longevity. Mice deficient in the p66Shc gene are long-lived, and their cells are both resistant to oxidative stress and produce less ROS. Our microarray analysis of p66Shc(-/-) mouse tissues showed alterations in transcripts involved in heme and superoxide production and insulin signaling. Thus, we carried out analysis of ROS production by NADPH oxidase (PHOX) in macrophages of control and p66Shc knock-out mice. p66Shc(-/-) mice had a 40% reduction in PHOX-dependent superoxide production. To confirm whether the defect in superoxide production was a direct consequence of p66Shc deficiency, p66Shc was knocked down with siRNA in the macrophage cell line RAW264, and a 30% defect in superoxide generation was observed. The pathway of PHOX-dependent superoxide generation was investigated. PHOX protein levels were not decreased in mutant macrophages; however, the rate and extent of phosphorylation of p47phox was decreased in mutants, as was membrane translocation of the complex. Consistently, phosphorylation of protein kinase Cdelta, Akt, and ERK (the kinases responsible for phosphorylation of p47phox) was decreased. Thus, p66Shc deficiency causes a defect in activation of the PHOX complex that results in decreased superoxide production. p66Shc-deficient mice have recently been observed to be resistant to atherosclerosis and to oxidant injury in kidney and brain. Because phagocyte-derived superoxide is often a component of oxidant injury and inflammation, we suggest that the decreased superoxide production by PHOX in p66Shc-deficient mice could contribute significantly to their relative protection from oxidant injury and consequent longevity.


Assuntos
Longevidade , NADPH Oxidases/metabolismo , Estresse Oxidativo , Proteínas Adaptadoras da Sinalização Shc , Superóxidos/metabolismo , Animais , Linhagem Celular , Ativação Enzimática/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Knockout , NADPH Oxidases/genética , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
14.
Toxicol Rep ; 8: 646-656, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33868951

RESUMO

Humans are frequently exposed to Quaternary Ammonium Compounds (QACs). QACs are ubiquitously used in medical settings, restaurants, and homes as cleaners and disinfectants. Despite their prevalence, nothing is known about the health effects associated with chronic low-level exposure. Chronic QAC toxicity, only recently identified in mice, resulted in developmental, reproductive, and immune dysfunction. Cell based studies indicate increased inflammation, decreased mitochondrial function, and disruption of cholesterol synthesis. If these findings translate to human toxicity, multiple physiological processes could be affected. This study tested whether QAC concentrations could be detected in the blood of 43 human volunteers, and whether QAC concentrations influenced markers of inflammation, mitochondrial function, and cholesterol synthesis. QAC concentrations were detected in 80 % of study participants. Blood QACs were associated with increase in inflammatory cytokines, decreased mitochondrial function, and disruption of cholesterol homeostasis in a dose dependent manner. This is the first study to measure QACs in human blood, and also the first to demonstrate statistically significant relationships between blood QAC and meaningful health related biomarkers. Additionally, the results are timely in light of the increased QAC disinfectant exposure occurring due to the SARS-CoV-2 pandemic. MAIN FINDINGS: This study found that 80 % of study participants contained QACs in their blood; and that markers of inflammation, mitochondrial function, and sterol homeostasis varied with blood QAC concentration.

15.
Aging (Albany NY) ; 13(6): 7914-7930, 2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33735837

RESUMO

Declines in mitochondrial mass are thought to be a hallmark of mammalian aging, and a ketogenic diet (KD) may prevent the age-related decreases in mitochondrial content. The objective of this study was to investigate the impact of a KD on markers of mitochondrial mass. Mice were fed an isocaloric control diet (CD) or KD from 12 months of age. Tissues were collected after 1 month and 14 months of intervention, and a panel of commonly used markers of mitochondrial mass (mitochondrial enzyme activities and levels, mitochondrial to nuclear DNA ratio, and cardiolipin content) were measured. Our results showed that a KD stimulated activities of marker mitochondrial enzymes including citrate synthase, Complex I, and Complex IV in hindlimb muscle in aged mice. KD also increased the activity of citrate synthase and prevented an age-related decrease in Complex IV activity in aged brain. No other markers were increased in these tissues. Furthermore, the impacts of a KD on liver and kidney were mixed with no pattern indicative of a change in mitochondrial mass. In conclusion, results of the present study suggest that a KD induces tissue-specific changes in mitochondrial enzyme activities, or structure, rather than global changes in mitochondrial mass across tissues.


Assuntos
Dieta Cetogênica , Rim/metabolismo , Fígado/metabolismo , Mitocôndrias/metabolismo , Animais , Complexo I de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Masculino , Camundongos
16.
Bioorg Med Chem ; 18(17): 6429-41, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20691600

RESUMO

Analogues of mitoQ and idebenone were synthesized to define the structural elements that support oxygen consumption in the mitochondrial respiratory chain. Eight analogues were prepared and fully characterized, then evaluated for their ability to support oxygen consumption in the mitochondrial respiratory chain. While oxygen consumption was strongly inhibited by mitoQ analogues 2-4 in a chain length-dependent manner, modification of idebenone by replacement of the quinone methoxy groups by methyl groups (analogues 6-8) reduced, but did not eliminate, oxygen consumption. Idebenone analogues 6-8 also displayed significant cytoprotective properties toward cultured mammalian cells in which glutathione had been depleted by treatment with diethyl maleate.


Assuntos
Mitocôndrias/efeitos dos fármacos , Compostos Organofosforados/síntese química , Compostos Organofosforados/farmacologia , Consumo de Oxigênio/efeitos dos fármacos , Ubiquinona/análogos & derivados , Antioxidantes/síntese química , Antioxidantes/farmacologia , Citoproteção , Glutationa/metabolismo , Humanos , Mitocôndrias/metabolismo , Ubiquinona/síntese química , Ubiquinona/farmacologia
17.
Mitochondrion ; 50: 19-24, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31654752

RESUMO

AMP-activated protein kinase (AMPK) is a eukaryotic energy sensor and protector from mitochondrial/energetic stress that is also a therapeutic target for cancer and metabolic disease. Metformin is an AMPK inducer that has been used in cancer therapeutic trials. Through screening we isolated cetylpyridinium chloride (CPC), a drug known to dose-dependently inhibit mitochondrial complex 1, as a potent and dose-dependent AMPK stimulator. Mitochondrial biogenesis and bioenergetics changes have also been implicated in glioblastoma, which is the most aggressive form of brain tumors. Cetylpyridinium chloride has been administered in humans as a safe drug-disinfectant for several decades, and we report here that under in vitro conditions, cetylpyridinium chloride kills glioblastoma cells in a dose dependent manner at a higher efficacy compared to current standard of care drug, temozolomide.


Assuntos
Adenilato Quinase/metabolismo , Antineoplásicos/farmacologia , Cetilpiridínio/farmacologia , Hepatócitos/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Anti-Infecciosos Locais/farmacologia , Linhagem Celular , Sobrevivência Celular , Glioma/tratamento farmacológico , Humanos , Camundongos
18.
Neurobiol Dis ; 34(2): 357-65, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19233273

RESUMO

Maternal inheritance of a pathogenic point mutation within complex I of the mitochondrial genome causes Leber's hereditary optic neuropathy (LHON), resulting in the neurodegeneration and demyelination of the optic nerve. The integrated stress response (ISR), a signaling pathway that responds to various stresses by activating a common set of genes, has been linked to both mitochondrial defects and demyelinating diseases. Therefore, we wanted to determine whether mitochondrial dysfunction induced by complex I inhibition with rotenone can activate the ISR, specifically by the ER kinase PERK, in oligodendroglial cells. Our complex I-deficient oligodendroglial model reproduced similar biochemical defects as in LHON by decreasing ATP synthesis and ATP levels. The same doses of rotenone that reduced ATP production also induced dose-dependent increases in PERK and eIF2alpha phosphorylation as well as activated the ISR stress genes, ATF4 and CHOP. In addition, complex I inhibition at these same concentrations induced a PERK-dependent activation of the cell death kinase, JNK, and inhibited oligodendroglial proliferation. Taken together, our results demonstrate that activation of the ISR may be one example of mitochondrial retrograde signaling in response to complex I deficiency and we suggest that this response mechanism may be relevant to the pathophysiology of LHON.


Assuntos
Complexo I de Transporte de Elétrons/genética , Mitocôndrias/metabolismo , Oligodendroglia/metabolismo , Atrofia Óptica Hereditária de Leber/metabolismo , Nervo Óptico/metabolismo , Estresse Fisiológico/genética , Fator 4 Ativador da Transcrição/efeitos dos fármacos , Fator 4 Ativador da Transcrição/metabolismo , Trifosfato de Adenosina/biossíntese , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Respiração Celular/efeitos dos fármacos , Respiração Celular/genética , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Humanos , Mitocôndrias/genética , Proteína Quinase 8 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Oligodendroglia/patologia , Atrofia Óptica Hereditária de Leber/genética , Atrofia Óptica Hereditária de Leber/fisiopatologia , Nervo Óptico/patologia , Nervo Óptico/fisiopatologia , Fator de Transcrição CHOP/efeitos dos fármacos , Fator de Transcrição CHOP/metabolismo , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Desacopladores/toxicidade , eIF-2 Quinase/efeitos dos fármacos , eIF-2 Quinase/metabolismo
19.
Anal Biochem ; 385(1): 176-8, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18950600

RESUMO

The measurement of mitochondrial activity in living cells is usually not straightforward, even though it is quite important in physiological and pathophysiological processes. We describe a high-throughput method for measurement of mitochondrial oxygen consumption in living cells, based on the Becton-Dickinson Biosensor plates.


Assuntos
Técnicas Biossensoriais/métodos , Proteínas Mitocondriais/análise , Proteínas Mitocondriais/genética , Mutação Puntual/genética , Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/farmacologia , Relação Dose-Resposta a Droga , Humanos , Proteínas Mitocondriais/metabolismo , Consumo de Oxigênio , Fatores de Tempo , Células Tumorais Cultivadas
20.
PLoS One ; 14(10): e0223209, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31665133

RESUMO

Friedreich's ataxia (FA) is a neurodegenerative disease with no approved therapy that is the result of frataxin deficiency. The identification of human FA blood biomarkers related to disease severity and neuro-pathomechanism could support clinical trials of drug efficacy. To try to identify human biomarkers of neuro-pathomechanistic relevance, we compared the overlapping gene expression changes of primary blood and skin cells of FA patients with changes in the Dorsal Root Ganglion (DRG) of the KIKO FA mouse model. As DRG is the primary site of neurodegeneration in FA, our goal was to identify which changes in blood and skin of FA patients provide a 'window' into the FA neuropathomechanism inside the nervous system. In addition, gene expression in frataxin-deficient neuroglial cells and FA mouse hearts were compared for a total of 5 data sets. The overlap of these changes strongly supports mitochondrial changes, apoptosis and alterations of selenium metabolism. Consistent biomarkers were observed, including three genes of mitochondrial stress (MTIF2, ENO2), apoptosis (DDIT3/CHOP), oxidative stress (PREX1), and selenometabolism (SEPW1). These results prompted our investigation of the GPX1 activity as a marker of selenium and oxidative stress, in which we observed a significant change in FA patients. We believe these lead biomarkers that could be assayed in FA patient blood as indicators of disease severity and progression, and also support the involvement of mitochondria, apoptosis and selenium in the neurodegenerative process.


Assuntos
Biomarcadores/sangue , Ataxia de Friedreich/sangue , Gânglios Espinais/metabolismo , Estresse Oxidativo/genética , Animais , Antioxidantes/metabolismo , Apoptose/genética , Modelos Animais de Doenças , Fatores de Iniciação em Eucariotos/sangue , Ataxia de Friedreich/genética , Ataxia de Friedreich/patologia , Gânglios Espinais/patologia , Regulação da Expressão Gênica/genética , Fatores de Troca do Nucleotídeo Guanina/sangue , Humanos , Proteínas de Ligação ao Ferro/genética , Camundongos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/sangue , Miocárdio/metabolismo , Selênio/metabolismo , Fator de Transcrição CHOP/sangue , Frataxina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA