Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Front Biosci (Landmark Ed) ; 27(12): 322, 2022 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-36624950

RESUMO

Coenzyme Q10 (CoQ10) deficiency is broadly divided into two types, primary and secondary. Primary CoQ10 deficiencies are relatively rare disorders resulting from mutations in genes directly involved in the CoQ10 biosynthetic pathway, and are not a subject of this article. Secondary CoQ10 disorders are relatively common, and may occur for a variety of reasons; these include mutations in genes not directly related to the synthetic pathway, oxidative stress induced reduction of CoQ10, and the effects of pharmacological agents such as statins. CoQ10 is of key importance in cell metabolism; in addition to its role in mitochondrial oxidative phosphorylation, it is a major endogenous antioxidant, and has a role in the metabolism of sulphides, lipids and amino acids. Given its importance in cell metabolism, it is unsurprising that secondary CoQ10 deficiency has been linked with a wide range of disorders. In this article, we have reviewed evidence of secondary CoQ10 deficiency in both common and less common disorders, and highlighted those disorders in which CoQ10 supplementation has been shown to be of significant clinical benefit.


Assuntos
Doenças Mitocondriais , Humanos , Doenças Mitocondriais/tratamento farmacológico , Doenças Mitocondriais/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Ubiquinona/farmacologia , Suplementos Nutricionais
2.
Adv Exp Med Biol ; 1286: 77-85, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33725346

RESUMO

Fibromyalgia is a common chronic pain condition of unknown aetiology, although mitochondrial dysfunction, oxidative stress, and inflammation have been implicated in the pathophysiology of this disorder. Treatment generally involves physiotherapy, anticonvulsants, and antidepressant therapy; however, the symptomatic relief conferred by these treatments can be very variable, and there is a need for additional therapeutic strategies. One such treatment which is gaining a lot of interest is the use of coenzyme Q10 (CoQ10) supplementation. The therapeutic efficacy associated with CoQ10 supplementation is thought to arise from the ability of supplementation to restore an underlying deficit in CoQ10 status which has been associated with fibromyalgia together with the ability of CoQ10 to improve mitochondrial activity, restore cellular antioxidant capacity, and ameliorate inflammation. This chapter outlines the evidence supporting the therapeutic utility of CoQ10 in the treatment of fibromyalgia.


Assuntos
Fibromialgia , Antioxidantes/metabolismo , Antioxidantes/uso terapêutico , Fibromialgia/tratamento farmacológico , Fibromialgia/metabolismo , Humanos , Mitocôndrias/metabolismo , Estresse Oxidativo , Ubiquinona/análogos & derivados
3.
Biochim Biophys Acta Mol Basis Dis ; 1867(6): 166100, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33549744

RESUMO

Very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency is the most common inborn long-chain fatty acid oxidation (FAO) disorder. VLCAD deficiency is characterized by distinct phenotypes. The severe phenotypes are potentially life-threatening and affect the heart or liver, with a comparatively milder phenotype characterized by myopathic symptoms. There is an unmet clinical need for effective treatment options for the myopathic phenotype. The molecular mechanisms driving the gradual decrease in mitochondrial function and associated alterations of muscle fibers are unclear. The peroxisome proliferator-activated receptor (PPAR) pan-agonist bezafibrate is a potent modulator of FAO and multiple other mitochondrial functions and has been proposed as a potential medication for myopathic cases of long-chain FAO disorders. In vitro experiments have demonstrated the ability of bezafibrate to increase VLCAD expression and activity. However, the outcome of small-scale clinical trials has been controversial. We found VLCAD deficient patient fibroblasts to have an increased oxidative stress burden and deranged mitochondrial bioenergetic capacity, compared to controls. Applying heat stress under fasting conditions to bezafibrate pretreated patient cells, caused a marked further increase of mitochondrial superoxide levels. Patient cells failed to maintain levels of the essential thiol peptide antioxidant glutathione and experienced a decrease in cellular viability. Our findings indicate that chronic PPAR activation is a plausible initiator of long-term pathogenesis in VLCAD deficiency. Our findings further implicate disruption of redox homeostasis as a key pathogenic mechanism in VLCAD deficiency and support the notion that a deranged thiol metabolism might be an important pathogenic factor in VLCAD deficiency.


Assuntos
Bezafibrato/farmacologia , Síndrome Congênita de Insuficiência da Medula Óssea/tratamento farmacológico , Metabolismo Energético , Fibroblastos/efeitos dos fármacos , Hipolipemiantes/farmacologia , Erros Inatos do Metabolismo Lipídico/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Doenças Mitocondriais/tratamento farmacológico , Doenças Musculares/tratamento farmacológico , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Síndrome Congênita de Insuficiência da Medula Óssea/metabolismo , Síndrome Congênita de Insuficiência da Medula Óssea/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Erros Inatos do Metabolismo Lipídico/metabolismo , Erros Inatos do Metabolismo Lipídico/patologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Doenças Musculares/metabolismo , Doenças Musculares/patologia , Estresse Oxidativo , Receptores Ativados por Proliferador de Peroxissomo/genética
4.
Antioxidants (Basel) ; 11(1)2021 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-35052506

RESUMO

The ageing brain is characterised by changes at the physical, histological, biochemical and physiological levels. This ageing process is associated with an increased risk of developing a number of neurological disorders, notably Alzheimer's disease and Parkinson's disease. There is evidence that mitochondrial dysfunction and oxidative stress play a key role in the pathogenesis of such disorders. In this article, we review the potential therapeutic role in these age-related neurological disorders of supplementary coenzyme Q10, a vitamin-like substance of vital importance for normal mitochondrial function and as an antioxidant. This review is concerned primarily with studies in humans rather than in vitro studies or studies in animal models of neurological disease. In particular, the reasons why the outcomes of clinical trials supplementing coenzyme Q10 in these neurological disorders is discussed.

5.
Neurochem Res ; 46(1): 131-139, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32306167

RESUMO

Organophosphate (OP) compounds are widely used as pesticides and herbicides and exposure to these compounds has been associated with both chronic and acute forms of neurological dysfunction including cognitive impairment, neurophysiological problems and cerebral ataxia with evidence of mitochondrial impairment being associated with this toxicity. In view of the potential mitochondrial impairment, the present study aimed to investigate the effect of exposure to commonly used OPs, dichlorvos, methyl-parathion (parathion) and chloropyrifos (CPF) on the cellular level of the mitochondrial electron transport chain (ETC) electron carrier, coenzyme Q10 (CoQ10) in human neuroblastoma SH-SY5Y cells. The effect of a perturbation in CoQ10 status was also evaluated on mitochondrial function and cell viability. A significant decreased (P < 0.0001) in neuronal cell viability was observed following treatment with all three OPs (100 µM), with dichlorvos appearing to be the most toxic to cells and causing an 80% loss of viability. OP treatment also resulted in a significant diminution in cellular CoQ10 status, with levels of this isoprenoid being decreased by 72% (P < 0.0001), 62% (P < 0.0005) and 43% (P < 0.005) of control levels following treatment with dichlorvos, parathion and CPF (50 µM), respectively. OP exposure was also found to affect the activities of the mitochondrial enzymes, citrate synthase (CS) and mitochondrial electron transport chain (ETC) complex II+III. Dichlorvos and CPF (50 µM) treatment significantly decreased CS activity by 38% (P < 0.0001) and 35% (P < 0.0005), respectively compared to control levels in addition to causing a 54% and 57% (P < 0.0001) reduction in complex II+III activity, respectively. Interestingly, although CoQ10 supplementation (5 µM) was able to restore cellular CoQ10 status and CS activity to control levels following OP treatment, complex II+III activity was only restored to control levels in neuronal cells exposed to dichlorvos (50 µM). However, post supplementation with CoQ10, complex II+III activity significantly increased by 33% (P < 0.0005), 25% (P < 0.005) and 35% (P < 0.0001) in dichlorvos, parathion and CPF (100 µM) treated cells respectively compared to non-CoQ10 supplemented cells. In conclusion, the results of this study have indicated evidence of neuronal cell CoQ10 deficiency with associated mitochondrial dysfunction following OP exposure. Although CoQ10 supplementation was able to ameliorate OP induced deficiencies in CS activity, ETC complex II+III activity appeared partially refractory to this treatment. Accordingly, these results indicate the therapeutic potential of CoQ10 supplementation in the treatment of OP poisoning. However, higher doses may be required to engender therapeutic efficacy.


Assuntos
Clorpirifos/toxicidade , Diclorvós/toxicidade , Inseticidas/toxicidade , Metil Paration/toxicidade , Neurônios/efeitos dos fármacos , Ubiquinona/análogos & derivados , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Complexo II de Transporte de Elétrons/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Ubiquinona/metabolismo , Ubiquinona/farmacologia
6.
J Equine Vet Sci ; 96: 103303, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33349408

RESUMO

Although coenzyme Q10 (CoQ10) serves as an antioxidant and energy source for spermatozoa when added to stallion semen before cooling or freezing, the effects of feeding CoQ10 on semen quality have not been studied. We assessed the effects of daily oral ingestion of CoQ10-ubiquinol by stallions on their plasma CoQ10 concentrations and semen quality. Seven mature Andalusian stallions ate 1g ubiquinol/day for 4 weeks followed by a 4-week washout period. Four horses initially completed an additional 4-week control period without ubiquinol. Blood was sampled weekly for determination of plasma CoQ10 concentrations. Ejaculates were collected every two weeks and assessed for total motility (TM), progressive motility (PM), and viability (V) after cooling for 24hours (T1), immediate cryopreservation (T2), and cryopreservation after 24hours cooling (T3). Ingesting ubiquinol resulted in an increase in plasma CoQ10 concentration (P < .001). Two weeks of CoQ10-ubiquinol resulted in improved V with all treatments (T1: P = .007; T2: P = .05; T3: P = .01) and PM with T3 (P = .04). In five stallions, TM and PM were also improved for T1 (P = .01 and P = .02, respectively) and TM increased with T2 (P = .03). Overall, semen quality parameters increased within the first 2 weeks of supplementation, plateaued at the end of the 4-week supplementation period and persisted after discontinuing ubiquinol until the end of the sampling period (8 weeks). Feeding 1 g CoQ10-ubiquinol for 4 weeks to breeding stallions improved semen quality after cooling and freezing in 5 of 7 stallions. This could be important for improving reproductive efficiency in stallions.


Assuntos
Análise do Sêmen , Preservação do Sêmen , Animais , Cavalos , Masculino , Plasma , Análise do Sêmen/veterinária , Preservação do Sêmen/veterinária , Ubiquinona/análogos & derivados
7.
Adv Exp Med Biol ; 1178: 103-112, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31493224

RESUMO

Coenzyme Q10 (CoQ10) is a vitamin-like substance which functions as an electron carrier within the mitochondrial respiratory chain, as well as serving as an important intracellular antioxidant. Most of the body's CoQ10 requirements are met by endogenous synthesis, although the capacity for CoQ10 production decreases substantially with increasing age. In this article we have reviewed the potential role of CoQ10 supplementation in the treatment of tissue fibrosis, which has been implicated in the age-related loss of function of various organs including the heart. Clinical studies have indicated that CoQ10 supplementation may decrease the level of cardiovascular fibrosis to which older individuals are subjected, and thereby improve cardiovascular function and reduce the risk of cardiovascular associated mortality. Although the factors responsible for the anti-fibrotic action of CoQ10 have yet to be fully elucidated, its antioxidant and anti-inflammatory functions are thought to be major contributors to its clinical efficacy in the treatment of this age-related disorder.


Assuntos
Antioxidantes , Ubiquinona/análogos & derivados , Antioxidantes/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/patologia , Fibrose/tratamento farmacológico , Humanos , Ubiquinona/uso terapêutico
8.
Mitochondrion ; 34: 103-114, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28263872

RESUMO

Spinocerebellar ataxia type 2 (SCA2) is a rare neurodegenerative disorder caused by a CAG repeat expansion in the ataxin-2 gene. We show increased oxidative stress, abnormalities in the antioxidant system, changes in complexes involved in oxidative phosphorylation and changes in mitochondrial morphology in SCA2 patient fibroblasts compared to controls, and we show that treatment with CoQ10 can partially reverse these changes. Together, our results suggest that oxidative stress and mitochondrial dysfunction may be contributory factors to the pathophysiology of SCA2 and that therapeutic strategies involving manipulation of the antioxidant system could prove to be of clinical benefit.


Assuntos
Fibroblastos/patologia , Mitocôndrias/patologia , Estresse Oxidativo , Ataxias Espinocerebelares/patologia , Ubiquinona/análogos & derivados , Vitaminas/metabolismo , Adolescente , Adulto , Idoso , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ubiquinona/metabolismo , Adulto Jovem
9.
Am J Clin Nutr ; 103(2): 579-88, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26718412

RESUMO

BACKGROUND: It is well established that low birth weight and accelerated postnatal growth increase the risk of liver dysfunction in later life. However, molecular mechanisms underlying such developmental programming are not well characterized, and potential intervention strategies are poorly defined. OBJECTIVES: We tested the hypotheses that poor maternal nutrition and accelerated postnatal growth would lead to increased hepatic fibrosis (a pathological marker of liver dysfunction) and that postnatal supplementation with the antioxidant coenzyme Q10 (CoQ10) would prevent this programmed phenotype. DESIGN: A rat model of maternal protein restriction was used to generate low-birth-weight offspring that underwent accelerated postnatal growth (termed "recuperated"). These were compared with control rats. Offspring were weaned onto standard feed pellets with or without dietary CoQ10 (1 mg/kg body weight per day) supplementation. At 12 mo, hepatic fibrosis, indexes of inflammation, oxidative stress, and insulin signaling were measured by histology, Western blot, ELISA, and reverse transcriptase-polymerase chain reaction. RESULTS: Hepatic collagen deposition (diameter of deposit) was greater in recuperated offspring (mean ± SEM: 12 ± 2 µm) than in controls (5 ± 0.5 µm) (P < 0.001). This was associated with greater inflammation (interleukin 6: 38% ± 24% increase; P < 0.05; tumor necrosis factor α: 64% ± 24% increase; P < 0.05), lipid peroxidation (4-hydroxynonenal, measured by ELISA: 0.30 ± 0.02 compared with 0.19 ± 0.05 µg/mL per µg protein; P < 0.05), and hyperinsulinemia (P < 0.05). CoQ10 supplementation increased (P < 0.01) hepatic CoQ10 concentrations and ameliorated liver fibrosis (P < 0.001), inflammation (P < 0.001), some measures of oxidative stress (P < 0.001), and hyperinsulinemia (P < 0.01). CONCLUSIONS: Suboptimal in utero nutrition combined with accelerated postnatal catch-up growth caused more hepatic fibrosis in adulthood, which was associated with higher indexes of oxidative stress and inflammation and hyperinsulinemia. CoQ10 supplementation prevented liver fibrosis accompanied by downregulation of oxidative stress, inflammation, and hyperinsulinemia.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Suplementos Nutricionais , Retardo do Crescimento Fetal/dietoterapia , Hepatite/prevenção & controle , Cirrose Hepática/prevenção & controle , Estresse Oxidativo , Ubiquinona/análogos & derivados , Animais , Citocinas/antagonistas & inibidores , Citocinas/sangue , Citocinas/metabolismo , Dieta com Restrição de Proteínas/efeitos adversos , Feminino , Desenvolvimento Fetal , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/imunologia , Retardo do Crescimento Fetal/fisiopatologia , Hepatite/etiologia , Hepatite/metabolismo , Hepatite/patologia , Hiperinsulinismo/etiologia , Hiperinsulinismo/prevenção & controle , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/etiologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Masculino , Desnutrição/fisiopatologia , Fenômenos Fisiológicos da Nutrição Materna , Gravidez , Complicações na Gravidez/fisiopatologia , Ratos Wistar , Organismos Livres de Patógenos Específicos , Ubiquinona/uso terapêutico , Desmame
10.
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
11.
J Neurol ; 261(11): 2192-8, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25182700

RESUMO

Inherited ataxias are heterogeneous disorders affecting both children and adults, with over 40 different causative genes, making molecular genetic diagnosis challenging. Although recent advances in next-generation sequencing have significantly improved mutation detection, few treatments exist for patients with inherited ataxia. In two patients with adult-onset cerebellar ataxia and coenzyme Q10 (CoQ10) deficiency in muscle, whole exome sequencing revealed mutations in ANO10, which encodes anoctamin 10, a member of a family of putative calcium-activated chloride channels, and the causative gene for autosomal recessive spinocerebellar ataxia-10 (SCAR10). Both patients presented with slowly progressive ataxia and dysarthria leading to severe disability in the sixth decade. Epilepsy and learning difficulties were also present in one patient, while retinal degeneration and cataract were present in the other. The detection of mutations in ANO10 in our patients indicate that ANO10 defects cause secondary low CoQ10 and SCAR10 patients may benefit from CoQ10 supplementation.


Assuntos
Ataxia/diagnóstico , Ataxia/genética , Proteínas de Membrana/genética , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/genética , Debilidade Muscular/diagnóstico , Debilidade Muscular/genética , Mutação/genética , Ubiquinona/deficiência , Adolescente , Adulto , Anoctaminas , Criança , Feminino , Humanos , Pessoa de Meia-Idade , Ubiquinona/genética , Adulto Jovem
12.
FASEB J ; 28(12): 5398-405, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25172893

RESUMO

Low birth weight and rapid postnatal growth increases risk of cardiovascular-disease (CVD); however, underlying mechanisms are poorly understood. Previously, we demonstrated that rats exposed to a low-protein diet in utero that underwent postnatal catch-up growth (recuperated) have a programmed deficit in cardiac coenzyme Q (CoQ) that was associated with accelerated cardiac aging. It is unknown whether this deficit occurs in all tissues, including those that are clinically accessible. We investigated whether aortic and white blood cell (WBC) CoQ is programmed by suboptimal early nutrition and whether postweaning dietary supplementation with CoQ could prevent programmed accelerated aging. Recuperated male rats had reduced aortic CoQ [22 d (35±8.4%; P<0.05); 12 m (53±8.8%; P<0.05)], accelerated aortic telomere shortening (P<0.01), increased DNA damage (79±13% increase in nei-endonucleaseVIII-like-1), increased oxidative stress (458±67% increase in NAPDH-oxidase-4; P<0.001), and decreased mitochondrial complex II-III activity (P<0.05). Postweaning dietary supplementation with CoQ prevented these detrimental programming effects. Recuperated WBCs also had reduced CoQ (74±5.8%; P<0.05). Notably, WBC CoQ levels correlated with aortic telomere-length (P<0.0001) suggesting its potential as a diagnostic marker of vascular aging. We conclude that early intervention with CoQ in at-risk individuals may be a cost-effective and safe way of reducing the global burden of CVDs.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Suplementos Nutricionais , Ubiquinona/metabolismo , Animais , Doenças Cardiovasculares/enzimologia , Feminino , Estresse Oxidativo , Gravidez , Ratos Wistar , Telomerase/metabolismo , Ubiquinona/administração & dosagem
13.
J Neurol Neurosurg Psychiatry ; 85(5): 493-8, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24218524

RESUMO

BACKGROUND: The autosomal-recessive cerebellar ataxias (ARCA) are a clinically and genetically heterogeneous group of neurodegenerative disorders. The large number of ARCA genes leads to delay and difficulties obtaining an exact diagnosis in many patients and families. Ubiquinone (CoQ10) deficiency is one of the potentially treatable causes of ARCAs as some patients respond to CoQ10 supplementation. The AarF domain containing kinase 3 gene (ADCK3) is one of several genes associated with CoQ10 deficiency. ADCK3 encodes a mitochondrial protein which functions as an electron-transfer membrane protein complex in the mitochondrial respiratory chain (MRC). METHODS: We report two siblings from a consanguineous Pakistani family who presented with cerebellar ataxia and severe myoclonus from adolescence. Whole exome sequencing and biochemical assessment of fibroblasts were performed in the index patient. RESULTS: A novel homozygous frameshift mutation in ADCK3 (p.Ser616Leufs*114), was identified in both siblings. This frameshift mutation results in the loss of the stop codon, extending the coding protein by 81 amino acids. Significant CoQ10 deficiency and reduced MRC enzyme activities in the index patient's fibroblasts suggested that the mutant protein may reduce the efficiency of mitochondrial electron transfer. CoQ10 supplementation was initiated following these genetic and biochemical analyses. She gained substantial improvement in myoclonic movements, ataxic gait and dysarthric speech after treatment. CONCLUSION: This study highlights the importance of diagnosing ADCK3 mutations and the potential benefit of treatment for patients. The identification of this new mutation broadens the phenotypic spectrum associated with ADCK3 mutations and provides further understanding of their pathogenic mechanism.


Assuntos
Ataxia Cerebelar/genética , Mutação da Fase de Leitura/genética , Proteínas Quinases/genética , Adulto , Ataxia Cerebelar/metabolismo , Ataxia Cerebelar/terapia , Consanguinidade , Feminino , Humanos , Proteínas Mitocondriais/genética , Linhagem , Ubiquinona/análogos & derivados , Ubiquinona/deficiência
14.
Mol Metab ; 2(4): 480-90, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24327963

RESUMO

Studies in human and animals have demonstrated that nutritionally induced low birth-weight followed by rapid postnatal growth increases the risk of metabolic syndrome and cardiovascular disease. Although the mechanisms underlying such nutritional programming are not clearly defined, increased oxidative-stress leading to accelerated cellular aging has been proposed to play an important role. Using an established rodent model of low birth-weight and catch-up growth, we show here that post-weaning dietary supplementation with coenzyme Q10, a key component of the electron transport chain and a potent antioxidant rescued many of the detrimental effects of nutritional programming on cardiac aging. This included a reduction in nitrosative and oxidative-stress, telomere shortening, DNA damage, cellular senescence and apoptosis. These findings demonstrate the potential for postnatal antioxidant intervention to reverse deleterious phenotypes of developmental programming and therefore provide insight into a potential translatable therapy to prevent cardiovascular disease in at risk humans.

15.
Neurochem Int ; 63(8): 750-5, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24140430

RESUMO

Deficiency of 5-methyltetrahydrofolate (5-MTHF) in cerebrospinal fluid (CSF) is associated with a number of neurometabolic conditions including mitochondrial electron transport chain defects. Whilst failure of the active transport of 5-methyltetrahydrofolate (5-MTHF) into the CSF compartment has been proposed as a potential mechanism responsible for the 5-MTHF deficiency seen in mitochondrial disorders, it is becoming increasingly clear that other mechanisms are involved. Here, we have considered the role of oxidative stress as a contributing mechanism. Concerning, ascorbic acid (AA), we have established a CSF reference range (103-303µM) and demonstrated a significant positive correlation between 5-MTHF and AA. Furthermore, CSF itself was also shown to convey antioxidant properties towards 5-MTHF. However, this protection could be overcome by the introduction of a hydroxyl radical generating system. Using a neuronal model system, inhibition of mitochondrial complex I, by 58%, was associated with a 23% increase in superoxide generation and a significantly increased loss of 5-MTHF from the extracellular medium. Addition of AA (150µM) was able to prevent this increased 5-MTHF catabolism. We conclude that increased generation of reactive oxygen species and/or loss of CSF antioxidants are also factors to consider with regard to the development of a central 5-MTHF deficiency. Co-supplementation of AA together with appropriate folate replacement may be of therapeutic benefit.


Assuntos
Ácido Ascórbico/líquido cefalorraquidiano , Ácido Fólico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Tetra-Hidrofolatos/líquido cefalorraquidiano , Adolescente , Adulto , Linhagem Celular Tumoral , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Adulto Jovem
16.
Mitochondrion ; 13(4): 337-41, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23583954

RESUMO

We evaluated coenzyme Q10 (CoQ) levels in patients studied under suspicion of mitochondrial DNA depletion syndromes (MDS) (n=39). CoQ levels were quantified by HPLC, and the percentage of mtDNA depletion by quantitative real-time PCR. A high percentage of MDS patients presented with CoQ deficiency as compared to other mitochondrial patients (Mann-Whitney-U test: p=0.001). Our findings suggest that MDS are frequently associated with CoQ deficiency, as a possible secondary consequence of disease pathophysiology. Assessment of muscle CoQ status seems advisable in MDS patients since the possibility of CoQ supplementation may then be considered as a candidate therapy.


Assuntos
Ataxia/epidemiologia , Erros Inatos do Metabolismo/complicações , Doenças Mitocondriais/complicações , Doenças Mitocondriais/epidemiologia , Miopatias Mitocondriais/complicações , Debilidade Muscular/epidemiologia , Doenças Musculares/complicações , Ubiquinona/deficiência , Adolescente , Ataxia/diagnóstico , Criança , Pré-Escolar , Cromatografia Líquida de Alta Pressão , DNA Mitocondrial/análise , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Doenças Mitocondriais/diagnóstico , Debilidade Muscular/diagnóstico , Reação em Cadeia da Polimerase em Tempo Real , Ubiquinona/análogos & derivados , Ubiquinona/análise , Adulto Jovem
17.
Rapid Commun Mass Spectrom ; 27(9): 924-30, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23592193

RESUMO

RATIONALE: Neurological dysfunction is common in primary coenzyme Q10 (2,3-dimethoxy, 5-methyl, 6-polyisoprene parabenzoquinone; CoQ10 ; ubiquinone) deficiencies, the most readily treatable subgroup of mitochondrial disorders. Therapeutic benefit from CoQ10 supplementation has also been noted in other neurodegenerative diseases. CoQ10 can be measured by high-performance liquid chromatography (HPLC) in plasma, muscle or leucocytes; however, there is no reliable method to quantify CoQ10 in cerebrospinal fluid (CSF). Additionally, many methods use CoQ9 , an endogenous ubiquinone in humans, as an internal standard. METHODS: Deuterated CoQ10 (d6 -CoQ10 ) was synthesised by a novel, simple, method. Total CoQ10 was measured by liquid chromatography/tandem mass spectrometry (LC/MS/MS) using d6 -CoQ10 as internal standard and 5 mM methylamine as an ion-pairing reagent. Chromatography was performed using a Hypsersil GOLD C4 column (150 × 3 mm, 3 µm). RESULTS: CoQ10 levels were linear over a concentration range of 0-200 nM (R(2) = 0.9995). The lower limit of detection was 2 nM. The inter-assay coefficient of variation (CV) was 3.6% (10 nM) and 4.3% (20 nM), and intra-assay CV 3.4% (10 nM) and 3.6% (20 nM). Reference ranges were established for CoQ10 in CSF (5.7-8.7 nM; n = 17), fibroblasts (57.0-121.6 pmol/mg; n = 50) and muscle (187.3-430.1 pmol/mg; n = 15). CONCLUSIONS: Use of d6 -CoQ10 internal standard has enabled the development of a sensitive LC/MS/MS method to accurately determine total CoQ10 levels. Clinical applications of CSF CoQ10 determination include identification of patients with cerebral CoQ10 deficiency, and monitoring CSF CoQ10 levels following supplementation.


Assuntos
Fibroblastos/química , Músculo Esquelético/química , Espectrometria de Massas em Tandem/métodos , Ubiquinona/análogos & derivados , Adolescente , Adulto , Células Cultivadas , Criança , Pré-Escolar , Cromatografia Líquida de Alta Pressão/métodos , Cromatografia Líquida de Alta Pressão/normas , Deutério/análise , Feminino , Humanos , Lactente , Limite de Detecção , Masculino , Pessoa de Meia-Idade , Oxirredução , Padrões de Referência , Espectrometria de Massas em Tandem/normas , Ubiquinona/análise , Ubiquinona/líquido cefalorraquidiano , Adulto Jovem
18.
Mol Genet Metab ; 89(3): 214-21, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16798039

RESUMO

We report a patient with relatively mild Leigh syndrome and mitochondrial respiratory chain complex II deficiency caused by a homozygous G555E mutation in the nuclear encoded flavoprotein subunit of succinate dehydrogenase. This mutation has previously been reported in a lethal-infantile presentation of complex II deficiency. Such marked phenotypic heterogeneity, although typical of heteroplasmic mutations in the mitochondrial genome, is unusual for nuclear mutations. Comparable activities and stability of mitochondrial respiratory chain enzymes were demonstrated in both patients, so other reasons for the phenotypic variability are considered.


Assuntos
Núcleo Celular/metabolismo , Complexo II de Transporte de Elétrons/genética , Doenças Mitocondriais/genética , Mutação/genética , Fenótipo , Sequência de Aminoácidos , Sequência de Bases , Criança , Pré-Escolar , Análise Mutacional de DNA , DNA Complementar/metabolismo , Complexo II de Transporte de Elétrons/química , Complexo II de Transporte de Elétrons/metabolismo , Eletroforese em Gel de Poliacrilamida , Humanos , Hipotálamo Médio/diagnóstico por imagem , Lactente , Imageamento por Ressonância Magnética , Masculino , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Radiografia , Ubiquinona/metabolismo
19.
Ann Clin Biochem ; 40(Pt 3): 207-18, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12803831

RESUMO

Coenzyme Q(10) (CoQ(10)) is the predominant form of ubiquinone in man. CoQ(10) functions as an electron carrier in the mitochondrial respiratory chain as well as serving as an important intracellular antioxidant. Lowered blood and tissue concentrations of CoQ(10) have been reported in a number of diseases, although whether this deficiency is the cause or an effect of the disease remains largely unresolved. Some studies have reported lowered plasma CoQ(10) concentrations after statin drug treatment of hypercholesterolaemic patients. However, a deficiency in CoQ(10) status has yet to be demonstrated in patients experiencing the rare myotoxic side-effects of these drugs. Most clinical investigations assessing the therapeutic potential of CoQ(10) have focused on cardiovascular disease, specifically congestive heart failure. Although a number of studies have reported clinical improvement in congestive heart failure patients after CoQ(10) supplementation to standard therapy, concerns about the design of these studies coupled to the small number of patients involved have limited their acceptance. Assessment of CoQ(10) status is generally based on plasma measurements. As plasma concentrations are influenced by a number of physiological factors and may not represent cellular concentrations, platelets, lymphocytes and fibroblasts may provide suitable alternatives for these measurements.


Assuntos
Colesterol/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo , Anticolesterolemiantes/efeitos adversos , Colesterol/sangue , Coenzimas , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Valores de Referência , Ubiquinona/química , Ubiquinona/deficiência , Ubiquinona/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA