Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 206
Filtrar
1.
Biomed Res Int ; 2020: 5246350, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32190670

RESUMO

PURPOSE: To explore the effects of depression on cardiac autonomic nerve function and related metabolic pathways, the heart rate variability (HRV) and urinary differential metabolites were detected on the college students with depression. METHODS: 12 female freshmen with depression were filtered by the Beck Depression Inventory (BDI-II) and Self-rating Depression Scale (SDS). By wearing an HRV monitoring system, time domain indexes and frequency domain indexes were measured over 24 hours. Liquid chromatography-mass spectrometry (LC-MS) was used to detect their urinary differential metabolites. Differential metabolites were identified by principal component analysis (PCA) and orthogonal projections to latent structures discriminant analysis (OPLS-DA). The metabolic pathways related to these differential metabolites were analyzed by the MetPA database. RESULTS: Stress time was significantly increased, and recovery time was markedly decreased in the depression group compared with the control group (p < 0.001). Standard deviation of the normal-to-normal R interval (SDNN), root mean square of the beat-to-beat differences (RMSSD), high frequency (HF), and low frequency (LF) were decreased significantly (p < 0.001). Standard deviation of the normal-to-normal R interval (SDNN), root mean square of the beat-to-beat differences (RMSSD), high frequency (HF), and low frequency (LF) were decreased significantly (. CONCLUSION: Some autonomic nervous system disruption, high stress, and poor fatigue recovery were confirmed in college students with depression. The metabolic mechanism involved the disruption of coenzyme Q biosynthesis, glycine-serine-threonine metabolism, tyrosine metabolism, pyrimidine metabolism, and steroid metabolism under daily stress.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Frequência Cardíaca/fisiologia , Ubiquinona/biossíntese , Adolescente , Depressão , Fadiga , Feminino , Glicina/metabolismo , Humanos , Metabolômica , Monitorização Fisiológica , Pirimidinas/metabolismo , Serina/metabolismo , Esteroides/metabolismo , Estresse Fisiológico , Estudantes , Treonina/metabolismo , Tirosina/metabolismo , Ubiquinona/fisiologia , Urina/química , Adulto Jovem
2.
Genetics ; 214(2): 381-395, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31852725

RESUMO

Endocrine-disrupting chemicals are ubiquitously present in our environment, but the mechanisms by which they adversely affect human reproductive health and strategies to circumvent their effects remain largely unknown. Here, we show in Caenorhabditis elegans that supplementation with the antioxidant Coenzyme Q10 (CoQ10) rescues the reprotoxicity induced by the widely used plasticizer and endocrine disruptor bisphenol A (BPA), in part by neutralizing DNA damage resulting from oxidative stress. CoQ10 significantly reduces BPA-induced elevated levels of germ cell apoptosis, phosphorylated checkpoint kinase 1 (CHK-1), double-strand breaks (DSBs), and chromosome defects in diakinesis oocytes. BPA-induced oxidative stress, mitochondrial dysfunction, and increased gene expression of antioxidant enzymes in the germline are counteracted by CoQ10. Finally, CoQ10 treatment also reduced the levels of aneuploid embryos and BPA-induced defects observed in early embryonic divisions. We propose that CoQ10 may counteract BPA-induced reprotoxicity through the scavenging of reactive oxygen species and free radicals, and that this natural antioxidant could constitute a low-risk and low-cost strategy to attenuate the impact on fertility by BPA.


Assuntos
Reparo do DNA/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ubiquinona/análogos & derivados , Animais , Antioxidantes/metabolismo , Compostos Benzidrílicos/farmacologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Dano ao DNA/fisiologia , Fertilidade/efeitos dos fármacos , Células Germinativas/metabolismo , Mutação em Linhagem Germinativa/genética , Mitocôndrias/metabolismo , Oócitos/metabolismo , Oxirredução , Fenóis/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Ubiquinona/metabolismo , Ubiquinona/fisiologia
3.
Crit Rev Food Sci Nutr ; 59(14): 2240-2257, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29451807

RESUMO

Coenzyme Q10 (CoQ10) is a ubiquitous molecule present in all eukaryotic organisms whose principal role in the cell is related to its participation in the electron transport chain in the inner mitochondrial membrane. CoQ10 plays a major role in the control of cell redox status, and both the amount and functionality of this molecule have been related to the regulation of reactive oxygen species generation. Numerous reports can be found discussing the implications of CoQ10 supplementation in human studies and clinical trials related to aging. However, few reviews have made an updating through the translational point of view to integrate both basic and clinical aspects. The aim of this paper is to review our current knowledge from CoQ10 implications at biochemical and physiological level, in order to unravel the molecular mechanisms involved in its application in clinical practice. Although the importance of CoQ10 has been mainly attributed to its role as an agent for energy transduction in mitochondria, new functions for CoQ10 have been described in the recent past years, including anti-inflammatory effects, gene expression regulation and lipid bilayer membranes stabilization, which explain its involvement in aging and age-related diseases such as cardiovascular diseases, renal failure and neurodegenerative diseases.


Assuntos
Envelhecimento/patologia , Ubiquinona/análogos & derivados , Vitaminas/fisiologia , Animais , Doenças Cardiovasculares/tratamento farmacológico , Humanos , Modelos Animais , Doenças Neurodegenerativas/tratamento farmacológico , Oxirredução , Insuficiência Renal Crônica/tratamento farmacológico , Ubiquinona/química , Ubiquinona/metabolismo , Ubiquinona/fisiologia , Ubiquinona/uso terapêutico
4.
Sci Rep ; 8(1): 14013, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30228311

RESUMO

Mitohormesis is an adaptive response induced by a mild mitochondrial stress that promotes longevity and metabolic health in different organisms. This mechanism has been proposed as the cause of the increase in the survival in Coq7+/- (Mclk1+/-) mice, which show hepatic reduction of COQ7, early mitochondrial dysfunction and increased oxidative stress. Our study shows that the lack of COQ9 in Coq9Q95X mice triggers the reduction of COQ7, COQ6 and COQ5, which results in an increase in life expectancy. However, our results reveal that the hepatic CoQ levels are not decreased and, therefore, neither mitochondrial dysfunction or increased oxidative stress are observed in liver of Coq9Q95X mice. These data point out the tissue specific differences in CoQ biosynthesis. Moreover, our results suggest that the effect of reduced levels of COQ7 on the increased survival in Coq9Q95X mice may be due to mitochondrial mechanisms in non-liver tissues or to other unknown mechanisms.


Assuntos
Longevidade , Mitocôndrias Hepáticas/metabolismo , Ubiquinona/biossíntese , Animais , Antioxidantes/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Hepáticas/patologia , Ubiquinona/fisiologia
5.
Essays Biochem ; 62(3): 377-398, 2018 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-30030365

RESUMO

Primary Coenzyme Q deficiencies represent a group of rare conditions caused by mutations in one of the genes required in its biosynthetic pathway at the enzymatic or regulatory level. The associated clinical manifestations are highly heterogeneous and mainly affect central and peripheral nervous system, kidney, skeletal muscle and heart. Genotype-phenotype correlations are difficult to establish, mainly because of the reduced number of patients and the large variety of symptoms. In addition, mutations in the same COQ gene can cause different clinical pictures. Here, we present an updated and comprehensive review of the clinical manifestations associated with each of the pathogenic variants causing primary CoQ deficiencies.


Assuntos
Ataxia/diagnóstico , Ataxia/genética , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/genética , Debilidade Muscular/diagnóstico , Debilidade Muscular/genética , Ubiquinona/análogos & derivados , Ubiquinona/deficiência , Ataxia/fisiopatologia , Ataxia/terapia , Genótipo , Humanos , Doenças Mitocondriais/fisiopatologia , Doenças Mitocondriais/terapia , Debilidade Muscular/fisiopatologia , Debilidade Muscular/terapia , Mutação , Fenótipo , Relação Estrutura-Atividade , Síndrome , Ubiquinona/biossíntese , Ubiquinona/química , Ubiquinona/genética , Ubiquinona/fisiologia
6.
Adv Nutr ; 9(4): 519S-523S, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30032220

RESUMO

3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) are extremely well tolerated but are associated with a range of mild-to-moderate statin-associated muscle symptoms (SAMS). Estimates of SAMS incidence vary from <1% in industry-funded clinical trials to 10-25% in nonindustry-funded clinical trials and ∼60% in some observational studies. SAMS are important because they result in dose reduction or discontinuation of these life-saving medications, accompanied by higher healthcare costs and cardiac events. The mechanisms that produce SAMS are not clearly defined. Statins block the production of farnesyl pyrophosphate, an intermediate in the mevalonate pathway, which is responsible for the production of coenzyme Q10 (CoQ10). This knowledge has prompted the hypothesis that reductions in plasma CoQ10 concentrations contribute to SAMS. Consequently, CoQ10 is popular as a form of adjuvant therapy for the treatment of SAMS. However, the data evaluating the efficacy of CoQ10 supplementation has been equivocal, with some, but not all, studies suggesting that CoQ10 supplementation mitigates muscular complaints. This review discusses the rationale for using CoQ10 in SAMS, the results of CoQ10 clinical trials, the suggested management of SAMS, and the lessons learned about CoQ10 treatment of this problem.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Doenças Musculares/induzido quimicamente , Doenças Musculares/tratamento farmacológico , Ubiquinona/análogos & derivados , Suplementos Nutricionais , Metabolismo Energético/fisiologia , Humanos , Músculo Esquelético/química , Doenças Musculares/genética , Mialgia/induzido quimicamente , Mialgia/tratamento farmacológico , Fosfatos de Poli-Isoprenil/antagonistas & inibidores , Polimorfismo de Nucleotídeo Único , Sesquiterpenos/antagonistas & inibidores , Ubiquinona/deficiência , Ubiquinona/fisiologia , Ubiquinona/uso terapêutico
7.
Cell Metab ; 28(1): 145-158.e4, 2018 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-29887397

RESUMO

Acute O2 sensing by peripheral chemoreceptors is essential for mammalian homeostasis. Carotid body glomus cells contain O2-sensitive ion channels, which trigger fast adaptive cardiorespiratory reflexes in response to hypoxia. O2-sensitive cells have unique metabolic characteristics that favor the hypoxic generation of mitochondrial complex I (MCI) signaling molecules, NADH and reactive oxygen species (ROS), which modulate membrane ion channels. We show that responsiveness to hypoxia progressively disappears after inducible deletion of the Ndufs2 gene, which encodes the 49 kDa subunit forming the coenzyme Q binding site in MCI, even in the presence of MCII substrates and chemical NAD+ regeneration. We also show contrasting effects of physiological hypoxia on mitochondrial ROS production (increased in the intermembrane space and decreased in the matrix) and a marked effect of succinate dehydrogenase activity on acute O2 sensing. Our results suggest that acute responsiveness to hypoxia depends on coenzyme QH2/Q ratio-controlled ROS production in MCI.


Assuntos
Corpo Carotídeo/metabolismo , Hipóxia/metabolismo , Canais Iônicos/metabolismo , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ubiquinona/fisiologia , Animais , Complexo I de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/metabolismo , Camundongos , NAD/metabolismo , NADH Desidrogenase/metabolismo
8.
Protein Sci ; 27(8): 1518-1525, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29675961

RESUMO

The young investigator award from the Protein Society was a special honor for me because, at its essence, the goal of my laboratory is to define what obscure proteins do. Years ago, I stumbled into mitochondria as a venue for this work, and these organelles continue to define the biological theme of my laboratory. Our approaches are fairly broad, reflecting my own somewhat unorthodox training among diverse scientific fields spanning organic synthesis, chemical biology, mechanistic biochemistry, signal transduction, and systems biology. Yet, whatever the theme or the discipline, we aim to understand how proteins work-especially those that hide in the dark corners of mitochondria. Below, I recount my own path into this arena of protein science, and describe how my experiences along the way have shaped our current multi-disciplinary efforts to define the inner workings of this complex biological system.


Assuntos
Biologia Computacional/métodos , Proteínas Mitocondriais , Animais , Humanos , Espectrometria de Massas , Camundongos , Proteínas Mitocondriais/química , Proteínas Mitocondriais/metabolismo , Proteínas Mitocondriais/fisiologia , Modelos Moleculares , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiologia , Ubiquinona/química , Ubiquinona/metabolismo , Ubiquinona/fisiologia
9.
Klin Monbl Augenheilkd ; 235(2): 157-162, 2018 Feb.
Artigo em Alemão | MEDLINE | ID: mdl-29448286

RESUMO

Mitochondrial function is closely linked to numerous aspects of eye health. Imbalance between the creation of energy and the development of reactive oxygen species (ROS) seems to be the cause of the development of mitochondrial dysfunctions. As a result of this energy deficit, the level of oxidative stress in the eye tissues increases, leading to numerous ophthalmic impairments. It is important to distinguish between primary mitochondrial eye diseases and secondary mitochondrial changes. Primary mitochondrial eye diseases, for example Leber's hereditary optic atrophy (LHON), retinitis pigmentosa and chronic progressive external ophthalmoplegia are caused by direct damage to mitochondrial function induced by defective genes, either located on mitochondrial DNA (mtDNA) or the DNA of the nucleus (nDNA). In contrast, secondary mitochondrial dysfunctions are caused by environmental factors. In recent years, there has been growing evidence that mitochondrial dysfunctions play an important role in many common eye diseases, such as glaucoma, dry eye, diabetic retinopathy, cataract and age-related macular degeneration (AMD). This article summarises current knowledge of mitochondrial dysfunctions and the role of coenzyme Q10 (CoQ10) as a possible treatment option - with a special focus on glaucoma.


Assuntos
Glaucoma/fisiopatologia , Doenças Mitocondriais/fisiopatologia , Ubiquinona/análogos & derivados , Animais , Disponibilidade Biológica , Diagnóstico Diferencial , Modelos Animais de Doenças , Transporte de Elétrons/fisiologia , Metabolismo Energético/fisiologia , Olho/fisiopatologia , Radicais Livres , Glaucoma/diagnóstico , Glaucoma/etiologia , Humanos , Microscopia Eletrônica , Doenças Mitocondriais/complicações , Doenças Mitocondriais/diagnóstico , Soluções Oftálmicas , Espécies Reativas de Oxigênio/metabolismo , Fatores de Risco , Ubiquinona/administração & dosagem , Ubiquinona/fisiologia
10.
Sci Rep ; 7(1): 17744, 2017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29255295

RESUMO

Primary ubiquinone (UQ) deficiency is an important subset of mitochondrial disease that is caused by mutations in UQ biosynthesis genes. To guide therapeutic efforts we sought to estimate the number of individuals who are born with pathogenic variants likely to cause this disorder. We used the NCBI ClinVar database and literature reviews to identify pathogenic genetic variants that have been shown to cause primary UQ deficiency, and used the gnomAD database of full genome or exome sequences to estimate the frequency of both homozygous and compound heterozygotes within seven genetically-defined populations. We used known population sizes to estimate the number of afflicted individuals in these populations and in the mixed population of the USA. We then performed the same analysis on predicted pathogenic loss-of-function and missense variants that we identified in gnomAD. When including only known pathogenic variants, our analysis predicts 1,665 affected individuals worldwide and 192 in the USA. Adding predicted pathogenic variants, our estimate grows to 123,789 worldwide and 1,462 in the USA. This analysis predicts that there are many undiagnosed cases of primary UQ deficiency, and that a large proportion of these will be in developing regions of the world.


Assuntos
Ataxia/epidemiologia , Ataxia/genética , Doenças Mitocondriais/epidemiologia , Doenças Mitocondriais/genética , Debilidade Muscular/epidemiologia , Debilidade Muscular/genética , Ubiquinona/deficiência , Bases de Dados de Ácidos Nucleicos , Exoma , Frequência do Gene , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Mutação/genética , Fenótipo , Ubiquinona/genética , Ubiquinona/fisiologia , Sequenciamento do Exoma
11.
J Biol Chem ; 292(49): 20086-20099, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29042439

RESUMO

Long-chain fatty acids (LCFAs) are used as a rich source of metabolic energy by several bacteria including important pathogens. Because LCFAs also induce oxidative stress, which may be detrimental to bacterial growth, it is imperative to understand the strategies employed by bacteria to counteract such stresses. Here, we performed a genetic screen in Escherichia coli on the LCFA, oleate, and compared our results with published genome-wide screens of multiple non-fermentable carbon sources. This large-scale analysis revealed that among components of the aerobic electron transport chain (ETC), only genes involved in the biosynthesis of ubiquinone, an electron carrier in the ETC, are highly required for growth in LCFAs when compared with other carbon sources. Using genetic and biochemical approaches, we show that this increased requirement of ubiquinone is to mitigate elevated levels of reactive oxygen species generated by LCFA degradation. Intriguingly, we find that unlike other ETC components whose requirement for growth is inversely correlated with the energy yield of non-fermentable carbon sources, the requirement of ubiquinone correlates with oxidative stress. Our results therefore suggest that a mechanism in addition to the known electron carrier function of ubiquinone is required to explain its antioxidant role in LCFA metabolism. Importantly, among the various oxidative stress combat players in E. coli, ubiquinone acts as the cell's first line of defense against LCFA-induced oxidative stress. Taken together, our results emphasize that ubiquinone is a key antioxidant during LCFA metabolism and therefore provides a rationale for investigating its role in LCFA-utilizing pathogenic bacteria.


Assuntos
Escherichia coli/metabolismo , Ácidos Graxos/metabolismo , Estresse Oxidativo , Ubiquinona/fisiologia , Antioxidantes , Escherichia coli/genética , Genoma Bacteriano , Ácido Oleico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ubiquinona/metabolismo
12.
Biomed Pharmacother ; 93: 17-26, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28622591

RESUMO

The aim of this study was to assess the possible modulatory effects of rosuvastatin and/or ubiquinone on trastuzumab (TRZ)-induced cardiotoxicity in mice. One hundred and twenty mice were divided into six equal groups as follows: control group; TRZ group; TRZ+carboxymethyl cellulose group; TRZ+rosuvastatin group; TRZ+Ubiquinone group and TRZ+rosuvastatin+Ubiquinone group. Serum creatine kinase (CK-MB), lactate dehydrogenase (LDH), troponin I and N-terminal pro-B-type natriuretic peptide (NT-pro BNP) were measured. Also, tissue malondialdehyde (MDA), catalase (CAT), glutathione peroxidase (GPx), interleukin 6 (IL-6), transforming growth factor beta 1 (TGF-ß1) and signal transducers and activators of transcription-3 (STAT-3) were determined. Also, echocardiography was performed. Parts of the heart were subjected to histopathological, immunohistochemical and electron microscopic examination. Administration of rosuvastatin and/or ubiquinone to TRZ-treated mice induced significant increase in tissue GPx, CAT and STAT-3 with significant decrease in serum CK-MB, LDH, troponin I, NT-pro BNP, tissue MDA, TGF-ß1 and IL-6 and improved the histopathological, immunohistochemical, echocardiographic and electron microscopic changes compared to the group that received TRZ alone. These changes were significant in rosuvastatin/ubiquinone combination group compared to the use of each of these drugs alone. In conclusion, rosuvastatin/ubiquinone combination may represent a new therapeutic modality to ameliorate TRZ-induced cardiotoxicity.


Assuntos
Citocinas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Rosuvastatina Cálcica/farmacologia , Fator de Transcrição STAT3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Trastuzumab/farmacologia , Ubiquinona/fisiologia , Animais , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais/efeitos dos fármacos
13.
J Card Fail ; 22(7): 548-59, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27002943

RESUMO

BACKGROUND: The mechanisms for cognitive impairment in heart failure (HF) are unclear. We investigated the relative contributions of cerebral blood flow velocity (BFV), oxidative stress, and inflammation to HF-associated cognitive impairment. METHODS AND RESULTS: Thirty-six HF patients (≥60 years) and 40 healthy controls (68 ± 7 vs 67 ± 5 years, P > .05; 69% vs 50% male, P > .05) completed the Cognitive Drug Research computerized assessment battery and Stroop tasks. Common carotid (CCA) and middle cerebral arterial BFV were obtained by transcranial Doppler. Blood samples were collected for oxidant (diacron-reactive oxygen metabolites; F2-isoprostanes), antioxidant (coenzyme Q10; CoQ10), and inflammatory markers (high-sensitivity C-reactive protein). Compared with controls, patients exhibited impaired attention (Cognitive Drug Research's Power of Attention domain, congruent Stroop) and executive function (incongruent Stroop). Multiple regression modeling showed that CCA-BFV and CoQ10 but not group predicted performance on attention and executive function. Additionally, in HF patients, CCA-BFV and CoQ10 (ß = -0.34 vs ß = -0.35) were significant predictors of attention, and CCA-BFV (ß = -0.34) was a predictor of executive function. CONCLUSIONS: Power of Attention and executive function is impaired in older HF patients, and reduced CCA-BFV and CoQ10 are associated with worse cognition. Interventions addressing these mechanisms may improve cognition in older HF patients.


Assuntos
Circulação Cerebrovascular/fisiologia , Transtornos Cognitivos/fisiopatologia , Cognição/fisiologia , Insuficiência Cardíaca/fisiopatologia , Inflamação/fisiopatologia , Estresse Oxidativo/fisiologia , Idoso , Velocidade do Fluxo Sanguíneo , Proteína C-Reativa , Transtornos Cognitivos/diagnóstico , Transtornos Cognitivos/etiologia , Feminino , Insuficiência Cardíaca/complicações , Humanos , Masculino , Pessoa de Meia-Idade , Testes Neuropsicológicos , Ubiquinona/fisiologia
14.
South Med J ; 109(1): 17-21, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26741866

RESUMO

Coenzyme Q-10 (CoQ10) is a widely used alternative medication or dietary supplement and one of its roles is as an antioxidant. It naturally functions as a coenzyme and component of oxidative phosphorylation in mitochondria. Decreased levels have been demonstrated in diseased myocardium and in Parkinson disease. Farnesyl pyrophosphate is a critical intermediate for CoQ10 synthesis and blockage of this step may be important in statin myopathy. Deficiency of CoQ10 also has been associated with encephalomyopathy, severe infantile multisystemic disease, cerebellar ataxia, nephrotic syndrome, and isolated myopathy. Although supplementation with CoQ10 has been reported to be beneficial in treating hypertension, congestive heart failure, statin myopathy, and problems associated with chemotherapy for cancer treatement, this use of CoQ10 as a supplement has not been confirmed in randomized controlled clinical trials. Nevertheless, it appears to be a safe supplementary medication where usage in selected clinical situations may not be inappropriate. This review is an attempt to actualize the available information on CoQ10 and define its potential benefit and appropriate usage.


Assuntos
Ubiquinona/análogos & derivados , Animais , Doenças Cardiovasculares/tratamento farmacológico , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Hipertensão/tratamento farmacológico , Doenças Musculares/induzido quimicamente , Doenças Musculares/tratamento farmacológico , Neoplasias/tratamento farmacológico , Ubiquinona/deficiência , Ubiquinona/fisiologia , Ubiquinona/uso terapêutico
15.
Nat Rev Nephrol ; 12(5): 267-80, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26804019

RESUMO

Mitochondria are increasingly recognized as key players in genetic and acquired renal diseases. Most mitochondrial cytopathies that cause renal symptoms are characterized by tubular defects, but glomerular, tubulointerstitial and cystic diseases have also been described. For example, defects in coenzyme Q10 (CoQ10) biosynthesis and the mitochondrial DNA 3243 A>G mutation are important causes of focal segmental glomerulosclerosis in children and in adults, respectively. Although they sometimes present with isolated renal findings, mitochondrial diseases are frequently associated with symptoms related to central nervous system and neuromuscular involvement. They can result from mutations in nuclear genes that are inherited according to classic Mendelian rules or from mutations in mitochondrial DNA, which are transmitted according to more complex rules of mitochondrial genetics. Diagnosis of mitochondrial disorders involves clinical characterization of patients in combination with biochemical and genetic analyses. In particular, prompt diagnosis of CoQ10 biosynthesis defects is imperative because of their potentially reversible nature. In acute kidney injury (AKI), mitochondrial dysfunction contributes to the physiopathology of tissue injury, whereas mitochondrial biogenesis has an important role in the recovery of renal function. Potential therapies that target mitochondrial dysfunction or promote mitochondrial regeneration are being developed to limit renal damage during AKI and promote repair of injured tissue.


Assuntos
Injúria Renal Aguda/fisiopatologia , Síndrome de Kearns-Sayre/fisiopatologia , Nefropatias/fisiopatologia , Mitocôndrias/fisiologia , Miopatias Mitocondriais/fisiopatologia , Alquil e Aril Transferases/genética , Animais , DNA Mitocondrial/genética , Humanos , Mutação , Fosforilação Oxidativa , Espécies Reativas de Oxigênio/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/deficiência , Ubiquinona/fisiologia
16.
Endocrinology ; 156(10): 3528-37, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26214037

RESUMO

Low birth weight and rapid postnatal growth increases the risk of developing insulin resistance and type 2 diabetes in later life. However, underlying mechanisms and potential intervention strategies are poorly defined. Here we demonstrate that male Wistar rats exposed to a low-protein diet in utero that had a low birth weight but then underwent postnatal catch-up growth (recuperated offspring) had reductions in the insulin signaling proteins p110-ß (13% ± 6% of controls [P < .001]) and insulin receptor substrate-1 (39% ± 10% of controls [P < .05]) in adipose tissue. These changes were not accompanied by any change in expression of the corresponding mRNAs, suggesting posttranscriptional regulation. Recuperated animals displayed evidence of a proinflammatory phenotype of their adipose tissue with increased IL-6 (139% ± 8% [P < .05]) and IL1-ß (154% ± 16% [P < .05]) that may contribute to the insulin signaling protein dysregulation. Postweaning dietary supplementation of recuperated animals with coenzyme Q (CoQ10) (1 mg/kg of body weight per day) prevented the programmed reduction in insulin receptor substrate-1 and p110-ß and the programmed increased in IL-6. These findings suggest that postweaning CoQ10 supplementation has antiinflammatory properties and can prevent programmed changes in insulin-signaling protein expression. We conclude that CoQ10 supplementation represents an attractive intervention strategy to prevent the development of insulin resistance that results from suboptimal in utero nutrition.


Assuntos
Inflamação/metabolismo , Resistência à Insulina , Insulina/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Transdução de Sinais , Ubiquinona/análogos & derivados , Tecido Adiposo/metabolismo , Animais , Feminino , Perfilação da Expressão Gênica , Transtornos do Crescimento/fisiopatologia , Insulina/sangue , Lipídeos/sangue , Masculino , Exposição Materna , Camundongos , MicroRNAs/metabolismo , Estresse Oxidativo , Fenótipo , Ratos , Ratos Wistar , Ubiquinona/fisiologia
17.
Eur J Clin Invest ; 45(7): 745-54, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25991405

RESUMO

BACKGROUND: Statins inhibit hydroxymethylglutaryl-coenzyme A reductase, decrease plasma low-density lipoprotein cholesterol and reduce cardiovascular morbidity and mortality. They can also exert adverse effects, mostly affecting skeletal muscle, ranging from mild myalgia to rhabdomyolysis. MATERIALS AND METHODS: Based on a PubMed search until December 2014, this review summarizes studies on statin effects on muscle mitochondrial morphology and function in the context of myopathy. RESULTS: Possible mechanisms of statin-induced myopathy include lower cholesterol synthesis and production of prenylated proteins, reduced dolichols and increased atrogin-1 expression. Statin-treated patients frequently feature decreased muscle coenzyme Q10 (CoQ10) contents, suggesting that statins might impair mitochondrial function. In cell cultures, statins diminish muscle oxygen consumption, promote mitochondrial permeability transient pore opening and generate apoptotic proteins. Animal models confirm the statin-induced decrease in muscle CoQ10, but reveal no changes in mitochondrial enzyme activities. Human studies yield contradictory results, with decreased CoQ10, elevated lipids, decreased enzyme activities in muscle and impaired maximal oxygen uptake in several but not all studies. Some patients are susceptible to statin-induced myopathy due to variations in genes encoding proteins involved in statin uptake and biotransformation such as the solute carrier organic anion transporter family member 1B1 (SLCO1B1) or cytochrome P450 (CYP2D6, CYP3A4, CYP3A5). Carriers for carnitine palmitoyltransferase II deficiency and McArdle disease also present with higher prevalence of statin-induced myopathy. CONCLUSIONS: Despite the widespread use of statins, the pathogenesis of statin-induced myopathy remains unclear, requiring prospective randomized controlled trials with intensive phenotyping also for identifying strategies for its risk assessment, prevention and treatment.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Mitocôndrias Musculares/fisiologia , Doenças Musculares/induzido quimicamente , Animais , Linhagem Celular , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Feminino , Humanos , Masculino , Mitocôndrias Musculares/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Coelhos , Ratos , Fatores de Risco , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo , Ubiquinona/fisiologia
18.
Annu Rev Nutr ; 35: 175-213, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25974695

RESUMO

Coenzyme Q (CoQ) is an essential lipid of cells present in all cellular compartments. The functions of CoQ in mitochondrial respiration and as an antioxidant are established, although the lipid likely has additional, presently unknown, roles. While the therapeutic utility of CoQ10 supplements is recognized in the rare cases of primary CoQ10 deficiencies, a potential role for CoQ10 supplements in cardiovascular disease, particularly heart failure, has also been studied for over 40 years. This review summarizes our current knowledge in these areas derived from animal studies and human trials. Current evidence for a benefit of CoQ10 supplements in diseases other than primary CoQ10 deficiencies is insufficient.


Assuntos
Insuficiência Cardíaca , Isquemia Miocárdica , Ubiquinona/análogos & derivados , Fatores Etários , Animais , Antioxidantes , Dieta , Suplementos Nutricionais , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Músculo Esquelético , Doenças Musculares/induzido quimicamente , Doenças Musculares/prevenção & controle , Isquemia Miocárdica/tratamento farmacológico , Distribuição Tecidual , Ubiquinona/administração & dosagem , Ubiquinona/deficiência , Ubiquinona/fisiologia
19.
Arterioscler Thromb Vasc Biol ; 34(9): 1860-70, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24675662

RESUMO

OBJECTIVE: Recent studies have shown the role of miRNAs in macrophage reverse cholesterol transport and atherogenesis. We hypothesized that coenzyme Q10 (CoQ10) may increase macrophage reverse cholesterol transport by regulating miRNA expression that contributes to the prevention of atherosclerosis. APPROACH AND RESULTS: CoQ10 treatment suppressed oxidized low-density lipoprotein-induced macrophage foam cell formation by ameliorating the binding of activator protein-1 to the putative promoter region of miR-378 primary transcript, thus decreasing the miR-378 level and enhancing the ATP-binding cassette transporter G1-mediated macrophage cholesterol efflux to high-density lipoprotein. Subsequently, the axis of activator protein-1/miR-378/ATP-binding cassette transporter G1 cholesterol efflux was confirmed in peritoneal macrophages isolated from CoQ10-treated apolipoprotein E-deficient mice. Finally, CoQ10 consumption promoted macrophage reverse cholesterol transport and inhibited the progression of atherosclerosis in apolipoprotein E-deficient mice. CONCLUSIONS: This study identified activator protein-1/miR-378/ATP-binding cassette transporter G1 as a novel cascade for CoQ10 in facilitating macrophage cholesterol efflux in vitro and in vivo. Our data thus imply that both CoQ10 and miR-378 are promising candidates for atherosclerosis prevention and treatment.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Aterosclerose/metabolismo , Colesterol/metabolismo , Lipoproteínas/fisiologia , Macrófagos/efeitos dos fármacos , MicroRNAs/fisiologia , Fator de Transcrição AP-1/fisiologia , Ubiquinona/análogos & derivados , Regiões 3' não Traduzidas , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Apolipoproteínas E/deficiência , Aterosclerose/genética , Aterosclerose/prevenção & controle , Linhagem Celular Tumoral , Células Espumosas/efeitos dos fármacos , Células Espumosas/metabolismo , Células HEK293 , Humanos , Lipoproteínas/genética , Lipoproteínas LDL/farmacologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Regiões Promotoras Genéticas , RNA Interferente Pequeno/farmacologia , Transfecção , Ubiquinona/farmacologia , Ubiquinona/fisiologia
20.
Neurobiol Aging ; 35(4): 847-57, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24246717

RESUMO

The aging risk factor for Parkinson's disease is described in terms of specific disease markers including mitochondrial and gene dysfunctions relevant to energy metabolism. This review details evidence for the ability of nutritional agents to manage these aging risk factors. The combination of alpha lipoic acid, acetyl-l-carnitine, coenzyme Q10, and melatonin supports energy metabolism via carbohydrate and fatty acid utilization, assists electron transport and adenosine triphosphate synthesis, counters oxidative and nitrosative stress, and raises defenses against protein misfolding, inflammatory stimuli, iron, and other endogenous or xenobiotic toxins. These effects are supported by gene expression via the antioxidant response element (ARE; Keap/Nrf2 pathway), and by peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1 alpha), a transcription coactivator, which regulates gene expression for energy metabolism and mitochondrial biogenesis, and maintains the structural integrity of mitochondria. The effectiveness and synergies of the combination against disease risks are discussed in relation to gene action, dopamine cell loss, and the accumulation and spread of pathology via misfolded alpha-synuclein. In addition there are potential synergies to support a neurorestorative role via glial derived neurotrophic factor expression.


Assuntos
Envelhecimento , Doença de Parkinson/etiologia , Doença de Parkinson/prevenção & controle , Acetilcarnitina/administração & dosagem , Acetilcarnitina/fisiologia , Trifosfato de Adenosina/biossíntese , Elementos de Resposta Antioxidante , Metabolismo dos Carboidratos , Transporte de Elétrons , Metabolismo Energético/genética , Ácidos Graxos/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Humanos , Inflamação/prevenção & controle , Melatonina/administração & dosagem , Melatonina/fisiologia , Mitocôndrias/genética , Óxido Nítrico/fisiologia , Estresse Oxidativo , Doença de Parkinson/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Deficiências na Proteostase/prevenção & controle , Fatores de Risco , Ácido Tióctico/administração & dosagem , Ácido Tióctico/fisiologia , Fatores de Transcrição , Ubiquinona/administração & dosagem , Ubiquinona/análogos & derivados , Ubiquinona/fisiologia , alfa-Sinucleína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA