Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Eur J Hum Genet ; 32(4): 426-434, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38316953

ABSTRACT

GEMIN5 exerts key biological functions regulating pre-mRNAs intron removal to generate mature mRNAs. A series of patients were reported harboring mutations in GEMIN5. No treatments are currently available for this disease. We treated two of these patients with oral Coenzyme Q10 (CoQ10), which resulted in neurological improvements, although MRI abnormalities remained. Whole Exome Sequencing demonstrated compound heterozygosity at the GEMIN5 gene in both cases: Case one: p.Lys742* and p.Arg1016Cys; Case two: p.Arg1016Cys and p.Ser411Hisfs*6. Functional studies in fibroblasts revealed a decrease in CoQ10 biosynthesis compared to controls. Supplementation with exogenous CoQ10 restored it to control intracellular CoQ10 levels. Mitochondrial function was compromised, as indicated by the decrease in oxygen consumption, restored by CoQ10 supplementation. Transcriptomic analysis of GEMIN5 patients compared with controls showed general repression of genes involved in CoQ10 biosynthesis. In the rigor mortis defective flies, CoQ10 levels were decreased, and CoQ10 supplementation led to an improvement in the adult climbing assay performance, a reduction in the number of motionless flies, and partial restoration of survival. Overall, we report the association between GEMIN5 dysfunction and CoQ10 deficiency for the first time. This association opens the possibility of oral CoQ10 therapy, which is safe and has no observed side effects after long-term therapy.


Subject(s)
Ataxia , Mitochondrial Diseases , Muscle Weakness , Ubiquinone , Ubiquinone/deficiency , Adult , Humans , Ubiquinone/genetics , Ubiquinone/therapeutic use , Ubiquinone/metabolism , Follow-Up Studies , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/genetics , Mutation , SMN Complex Proteins/genetics
2.
Brain ; 146(10): 4191-4199, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37170631

ABSTRACT

COQ7 encodes a hydroxylase responsible for the penultimate step of coenzyme Q10 (CoQ10) biosynthesis in mitochondria. CoQ10 is essential for multiple cellular functions, including mitochondrial oxidative phosphorylation, lipid metabolism, and reactive oxygen species homeostasis. Mutations in COQ7 have been previously associated with primary CoQ10 deficiency, a clinically heterogeneous multisystemic mitochondrial disorder. We identified COQ7 biallelic variants in nine families diagnosed with distal hereditary motor neuropathy with upper neuron involvement, expending the clinical phenotype associated with defects in this gene. A recurrent p.Met1? change was identified in five families from Brazil with evidence of a founder effect. Fibroblasts isolated from patients revealed a substantial depletion of COQ7 protein levels, indicating protein instability leading to loss of enzyme function. High-performance liquid chromatography assay showed that fibroblasts from patients had reduced levels of CoQ10, and abnormal accumulation of the biosynthetic precursor DMQ10. Accordingly, fibroblasts from patients displayed significantly decreased oxygen consumption rates in patients, suggesting mitochondrial respiration deficiency. Induced pluripotent stem cell-derived motor neurons from patient fibroblasts showed significantly increased levels of extracellular neurofilament light protein, indicating axonal degeneration. Our findings indicate a molecular pathway involving CoQ10 biosynthesis deficiency and mitochondrial dysfunction in patients with distal hereditary motor neuropathy. Further studies will be important to evaluate the potential benefits of CoQ10 supplementation in the clinical outcome of the disease.


Subject(s)
Mitochondrial Diseases , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Motor Neurons/metabolism , Mutation/genetics , Ubiquinone/genetics
3.
Ophthalmic Genet ; 44(5): 486-490, 2023 10.
Article in English | MEDLINE | ID: mdl-36420660

ABSTRACT

PURPOSE: To describe a case of primary coenzyme Q10 deficiency in a child manifesting as early-onset renal failure, retinal dystrophy, and optic atrophy leading to progressive vision loss. METHODS: Clinical presentation and workup including visual fields, electroretinogram, and optical coherence tomography are presented. Genetic testing was performed. RESULTS: An eight-year-old female with nephropathy requiring renal transplantation subsequently developed progressive cone-rod dystrophy and optic atrophy. The patient had negative results on a targeted next-generation sequencing retinal dystrophy panel but whole-exome sequencing revealed two variants in COQ2 (likely biallelic), consistent with a diagnosis of primary coenzyme Q10 deficiency. CONCLUSIONS: Primary coenzyme Q10 deficiency is a rare disorder with variable systemic and ocular findings; there is also genetic heterogeneity. Genetic testing aids in the diagnosis of this condition, and variants in the COQ2 and PDSS1 genes appear to have the strongest association with ocular manifestations. Oral supplementation of coenzyme Q10 may slow progression of disease. This case highlights the utility of whole-exome sequencing in the diagnosis of a rare syndromic form of ocular disease and reports a novel phenotypic association for this condition.


Subject(s)
Optic Atrophy , Retinal Dystrophies , Child , Female , Humans , Ubiquinone/therapeutic use , Ubiquinone/genetics , Genetic Testing , Retinal Dystrophies/genetics , Visual Fields , Electroretinography , Optic Atrophy/genetics , Mutation , Tomography, Optical Coherence
4.
Arch Insect Biochem Physiol ; 110(4): e21900, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35365940

ABSTRACT

Long-chain polyprenyl diphosphate synthases play a critical role in the formation of the prenyl side-chain of ubiquinones, but up to date, their functions have scarcely been characterized in insects. Here, we first cloned the complementary DNAs encoding the subunits of decaprenyl diphosphate synthase (DPPS) in the vetch aphid Megoura viciae, an important agricultural pest insect. The results showed that there existed three DPPS subunits, designated as MvDPPS1, MvDPPS2a, and MvDPPS2b, with an open reading frame of 1218, 1275, and 1290 bp, and a theoretical isoelectric point of 7.91, 6.63, and 9.62, respectively. The sequences of MvDPPS1s from different aphid species were nearly identical, while the sequences of MvDPPS2a and MvDPPS2b shared only moderate sequence similarity. Phylogenetic analysis clearly separated MvDPPS2a and MvDPPS2b, indicating a functional differentiation between them. Functional coexpression analysis in Escherichia coli showed that MvDPPS1 plus MvDPPS2a and MvDPPS1 plus MvDPPS2b, respectively, catalyzed the formation of the prenyl side-chain of the ubiquinone coenzyme Q10 (CoQ10). Interestingly, MvDPPS1 plus MvDPPS2b catalyzed the formation of the prenyl side-chain of a ubiquinone other than CoQ10. RNA interference-mediated knockdown of MvDPPS2a imposed no significant effect on MvDPPS2b, and vice versa, suggesting no compensatory action between them. In the end, we detected the product CoQ10 in the aphid, the first identification of CoQ10 in an insect species. Taken together, we characterized two functional DPPSs in M. viciae, one of which might be multifunctional. Our study helps to understand the functional plasticity of the terpenoid backbone biosynthesis pathway in insects.


Subject(s)
Alkyl and Aryl Transferases , Aphids , Vicia , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Animals , Aphids/genetics , Aphids/metabolism , Diphosphates/metabolism , Escherichia coli/genetics , Phylogeny , Ubiquinone/genetics , Ubiquinone/metabolism , Vicia/metabolism
5.
J Med Genet ; 59(9): 878-887, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34656997

ABSTRACT

BACKGROUND: Human coenzyme Q4 (COQ4) is essential for coenzyme Q10 (CoQ10) biosynthesis. Pathogenic variants in COQ4 cause childhood-onset neurodegeneration. We aimed to delineate the clinical spectrum and the cellular consequences of COQ4 deficiency. METHODS: Clinical course and neuroradiological findings in a large cohort of paediatric patients with COQ4 deficiency were analysed. Functional studies in patient-derived cell lines were performed. RESULTS: We characterised 44 individuals from 36 families with COQ4 deficiency (16 newly described). A total of 23 different variants were identified, including four novel variants in COQ4. Correlation analyses of clinical and neuroimaging findings revealed three disease patterns: type 1: early-onset phenotype with neonatal brain anomalies and epileptic encephalopathy; type 2: intermediate phenotype with distinct stroke-like lesions; and type 3: moderate phenotype with non-specific brain pathology and a stable disease course. The functional relevance of COQ4 variants was supported by in vitro studies using patient-derived fibroblast lines. Experiments revealed significantly decreased COQ4 protein levels, reduced levels of cellular CoQ10 and elevated levels of the metabolic intermediate 6-demethoxyubiquinone. CONCLUSION: Our study describes the heterogeneous clinical presentation of COQ4 deficiency and identifies phenotypic subtypes. Cell-based studies support the pathogenic characteristics of COQ4 variants. Due to the insufficient clinical response to oral CoQ10 supplementation, alternative treatment strategies are warranted.


Subject(s)
Mitochondrial Proteins , Ubiquinone , Cell Line , Child , Humans , Infant, Newborn , Mitochondrial Proteins/genetics , Neuroimaging , Phenotype , Ubiquinone/genetics , Ubiquinone/metabolism
6.
J Biol Chem ; 297(5): 101283, 2021 11.
Article in English | MEDLINE | ID: mdl-34626646

ABSTRACT

Ubiquinone (Coenzyme Q) is a vital respiratory cofactor and liposoluble antioxidant. In plants, it is not known how the C-6 hydroxylation of demethoxyubiquinone, the penultimate step in ubiquinone biosynthesis, is catalyzed. The combination of cross-species gene network modeling along with mining of embryo-defective mutant databases of Arabidopsis thaliana identified the embryo lethal locus EMB2421 (At1g24340) as a top candidate for the missing plant demethoxyubiquinone hydroxylase. In marked contrast with prototypical eukaryotic demethoxyubiquinone hydroxylases, the catalytic mechanism of which depends on a carboxylate-bridged di-iron domain, At1g24340 is homologous to FAD-dependent oxidoreductases that instead use NAD(P)H as an electron donor. Complementation assays in Saccharomyces cerevisiae and Escherichia coli demonstrated that At1g24340 encodes a functional demethoxyubiquinone hydroxylase and that the enzyme displays strict specificity for the C-6 position of the benzoquinone ring. Laser-scanning confocal microscopy also showed that GFP-tagged At1g24340 is targeted to mitochondria. Silencing of At1g24340 resulted in 40 to 74% decrease in ubiquinone content and de novo ubiquinone biosynthesis. Consistent with the role of At1g24340 as a benzenoid ring modification enzyme, this metabolic blockage could not be bypassed by supplementation with 4-hydroxybenzoate, the immediate precursor of ubiquinone's ring. Unlike in yeast, in Arabidopsis overexpression of demethoxyubiquinone hydroxylase did not boost ubiquinone content. Phylogenetic reconstructions indicated that plant demethoxyubiquinone hydroxylase is most closely related to prokaryotic monooxygenases that act on halogenated aromatics and likely descends from an event of horizontal gene transfer between a green alga and a bacterium.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Mitochondria , Mixed Function Oxygenases , Phylogeny , Ubiquinone , Arabidopsis/enzymology , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Mitochondria/enzymology , Mitochondria/genetics , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Ubiquinone/genetics , Ubiquinone/metabolism
7.
Metab Eng ; 68: 86-93, 2021 11.
Article in English | MEDLINE | ID: mdl-34555495

ABSTRACT

Coenzyme Q (CoQ) is vital for energy metabolism in living organisms. In humans, CoQ10 deficiency causes diseases and must be replenished via diet; however, CoQ content in plant foods is primarily low. Here, we report the breeding of high CoQ10 tomato lines by expressing four enzymes with a fruit-specific promoter, which modifies the chloroplast chorismate pathway, enhances cytosolic isoprenoid biosynthesis, and up-regulates the first two reactions in mitochondrion that construct the CoQ10 polyisoprenoid tail. We show that, while the level of the aromatic precursor could be markedly elevated, head group prenylation is the key to increasing the final CoQ10 yield. In the HUCD lines expressing all four transgenes, the highest CoQ10 content (0.15 mg/g dry weight) shows a seven-fold increase from the wild-type level and reaches an extraordinarily rich CoQ10 food grade. Overviewing the changes in other terpenoids by transcriptome and metabolic analyses reveals variable contents of carotenoids and α-tocopherol in the HUCD lines. In addition to the enigmatic relations among different terpenoid pathways, high CoQ10 plants maintaining substantial levels of either vitamin can be selected. Our investigation paves the way for the development of CoQ10-enriched crops as dietary supplements.


Subject(s)
Solanum lycopersicum , Ubiquinone , Carotenoids/metabolism , Fruit/metabolism , Humans , Solanum lycopersicum/genetics , Mitochondria , Ubiquinone/genetics
8.
Nanomedicine ; 37: 102439, 2021 10.
Article in English | MEDLINE | ID: mdl-34256063

ABSTRACT

Depletion of coenzyme Q (CoQ) is associated with disease, ranging from myopathy to heart failure. To induce a CoQ deficit, C2C12 myotubes were incubated with high dose simvastatin. This resulted in a concentration-dependent inhibition of cell viability. Simvastatin-induced effects were prevented by co-incubation with mevalonic acid. When myotubes were incubated with 60 µM simvastatin, mitochondrial CoQ content decreased while co-incubation with CoQ nanodisks (ND) increased mitochondrial CoQ levels and improved cell viability. Incubation of myotubes with simvastatin also led to a reduction in oxygen consumption rate (OCR). When myotubes were co-incubated with simvastatin and CoQ ND, the decline in OCR was ameliorated. The data indicate that CoQ ND represent a water soluble vehicle capable of delivering CoQ to cultured myotubes. Thus, these biocompatible nanoparticles have the potential to bypass poor CoQ oral bioavailability as a treatment option for individuals with severe CoQ deficiency syndromes and/or aging-related CoQ depletion.


Subject(s)
Ataxia/drug therapy , Mitochondrial Diseases/drug therapy , Muscle Weakness/drug therapy , Nanocomposites/chemistry , Simvastatin/adverse effects , Ubiquinone/deficiency , Ubiquinone/pharmacology , Animals , Ataxia/pathology , Cell Line , Cell Survival/drug effects , Heart Failure/chemically induced , Heart Failure/drug therapy , Heart Failure/pathology , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/pathology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Weakness/pathology , Muscular Diseases/chemically induced , Muscular Diseases/drug therapy , Muscular Diseases/pathology , Oxygen Consumption/drug effects , Simvastatin/pharmacology , Ubiquinone/chemistry , Ubiquinone/genetics
9.
Crit Rev Biochem Mol Biol ; 56(4): 360-372, 2021 08.
Article in English | MEDLINE | ID: mdl-33823724

ABSTRACT

Electron transfer flavoprotein dehydrogenase, also called ETF-ubiquinone oxidoreductase (ETF-QO), is a protein localized in the inner membrane of mitochondria, playing a central role in the electron-transfer system. Indeed, ETF-QO mediates electron transport from flavoprotein dehydrogenases to the ubiquinone pool. ETF-QO mutations are often associated with riboflavin-responsive multiple acyl-CoA dehydrogenase deficiency (RR-MADD, OMIM#231680), a multisystem genetic disease characterized by various clinical manifestations with different degrees of severity. In this review, we outline the clinical features correlated with ETF-QO deficiency and the benefits obtained from different treatments, such as riboflavin, L-carnitine and/or coenzyme Q10 supplementation, and a diet poor in fat and protein. Moreover, we provide a detailed summary of molecular and bioinformatic investigations, describing the mutations identified in ETFDH gene and highlighting their predicted impact on enzymatic structure and activity. In addition, we report biochemical and functional analysis, performed in HEK293 cells and patient fibroblasts and muscle cells, to show the relationship between the nature of ETFDH mutations, the variable impairment of enzyme function, and the different degrees of RR-MADD severity. Finally, we describe in detail 5 RR-MADD patients carrying different ETFDH mutations and presenting variable degrees of clinical symptom severity.


Subject(s)
Electron-Transferring Flavoproteins , Iron-Sulfur Proteins , Mitochondria , Multiple Acyl Coenzyme A Dehydrogenase Deficiency , Mutation , Oxidoreductases Acting on CH-NH Group Donors , Animals , Carnitine/genetics , Carnitine/metabolism , Electron-Transferring Flavoproteins/genetics , Electron-Transferring Flavoproteins/metabolism , Humans , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Mitochondria/enzymology , Mitochondria/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/enzymology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/genetics , Ubiquinone/metabolism
10.
Biofactors ; 47(4): 551-569, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33878238

ABSTRACT

Coenzyme Q10 (CoQ10 ) deficiency is a rare disease characterized by a decreased accumulation of CoQ10 in cell membranes. Considering that CoQ10 synthesis and most of its functions are carried out in mitochondria, CoQ10 deficiency cases are usually considered a mitochondrial disease. A relevant feature of CoQ10 deficiency is that it is the only mitochondrial disease with a successful therapy available, the CoQ10 supplementation. Defects in components of the synthesis machinery caused by mutations in COQ genes generate the primary deficiency of CoQ10 . Mutations in genes that are not directly related to the synthesis machinery cause secondary deficiency. Cases of CoQ10 deficiency without genetic origin are also considered a secondary deficiency. Both types of deficiency can lead to similar clinical manifestations, but the knowledge about primary deficiency is deeper than secondary. However, secondary deficiency cases may be underestimated since many of their clinical manifestations are shared with other pathologies. This review shows the current state of secondary CoQ10 deficiency, which could be even more relevant than primary deficiency for clinical activity. The analysis covers the fundamental features of CoQ10 deficiency, which are necessary to understand the biological and clinical differences between primary and secondary CoQ10 deficiencies. Further, a more in-depth analysis of CoQ10 secondary deficiency was undertaken to consider its origins, introduce a new way of classification, and include aging as a form of secondary deficiency.


Subject(s)
Aging/genetics , Alkyl and Aryl Transferases/genetics , Ataxia/genetics , GTP Phosphohydrolases/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Muscle Weakness/genetics , Niemann-Pick Disease, Type C/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Aging/metabolism , Alkyl and Aryl Transferases/metabolism , Animals , Ataxia/metabolism , Ataxia/pathology , Energy Metabolism/genetics , GTP Phosphohydrolases/metabolism , Gene Expression Regulation , Humans , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/metabolism , Muscle Weakness/metabolism , Muscle Weakness/pathology , Mutation , Niemann-Pick C1 Protein/genetics , Niemann-Pick C1 Protein/metabolism , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , Signal Transduction , Ubiquinone/genetics , Ubiquinone/metabolism
11.
Neurogenetics ; 22(1): 81-86, 2021 03.
Article in English | MEDLINE | ID: mdl-32830305

ABSTRACT

The spectrum of coenzyme Q10 (CoQ10) deficiency syndromes comprises a variety of disorders, including a form of autosomal recessive cerebellar ataxia (ARCA2) caused by mutations in the AarF domain-containing kinase 3 gene (ADCK3). Due to the potential response to CoQ10 supplementation, a timely diagnosis is crucial. Herein, we describe two siblings with a novel homozygous ADCK3 variant and an unusual presentation consisting of isolated writer's cramp with adult-onset. Cerebellar ataxia developed later in the disease course and remained stable during the follow-up. This report highlights that ARCA2 should be considered in the differential diagnosis of familial writer's cramp.


Subject(s)
Dystonic Disorders/genetics , Mutation/genetics , Ubiquinone/analogs & derivatives , Adult , Ataxia/genetics , Cerebellar Ataxia/diagnosis , Cerebellar Ataxia/genetics , Dystonic Disorders/diagnosis , Female , Homozygote , Humans , Mitochondrial Proteins/genetics , Ubiquinone/deficiency , Ubiquinone/genetics , Ubiquinone/metabolism
12.
Bratisl Lek Listy ; 121(11): 775-778, 2020.
Article in English | MEDLINE | ID: mdl-33164536

ABSTRACT

COVID-19 ‒ a coronavirus disease, affected almost all countries in the world. It is a new virus disease, nobody has prior immunity to it, human population is prone to infections. In March 11 2020, WHO declared the pandemic status. The main symptoms include: fever, dry cough and fatigue. Virus proteins need mitochondrial energy for their own survival and replication. Upon viral infections, mitochondrial dynamics and metabolism can be modulated, which can influence the energy production in the host cells. Coenzyme Q10 is an integral component of mitochondrial respiratory chain and the key component of mitochondrial ATP production. The exact pathobiochemical mechanism of the disease is unknown. Modulated mitochondrial dynamics and metabolism with lower CoQ10 levels in viral infections leads us to the hypothesis that one of the main pathobiochemical effects of SARS-Cov-2 virus could be mitochondrial bioenergetics dysfunction with CoQ10 deficit leading to the reduction of its endogenous biosynthesis. The mechanism might be virus induced oxidative stress causing a mutation of one or more of the nine COQ genes, resulting in primary CoQ10 deficiency. New perspective for patients with COVID-19 may be supportive targeting therapy with coenzyme Q10 to increase the energy production, immunity and decrease oxidative stress (Fig. 1, Ref. 51). Keywords: COVID-19, virus, mitochondrial bioenergetics, coenzyme Q10, oxidative stress.


Subject(s)
Coronavirus Infections/enzymology , Energy Metabolism , Mitochondria/enzymology , Pneumonia, Viral/enzymology , Ubiquinone/analogs & derivatives , Betacoronavirus , COVID-19 , Humans , Pandemics , SARS-CoV-2 , Ubiquinone/genetics
13.
PLoS One ; 15(11): e0242616, 2020.
Article in English | MEDLINE | ID: mdl-33232355

ABSTRACT

Coenzyme Q (CoQ, ubiquinone) is an essential component of the electron transport system in aerobic organisms. Human type CoQ10, which has 10 units of isoprene in its quinone structure, is especially valuable as a food supplement. Therefore, studying the biosynthesis of CoQ10 is important not only for increasing metabolic knowledge, but also for improving biotechnological production. Herein, we show that Schizosaccharomyces pombe utilizes p-aminobenzoate (PABA) in addition to p-hydroxybenzoate (PHB) as a precursor for CoQ10 synthesis. We explored compounds that affect the synthesis of CoQ10 and found benzoic acid (Bz) at >5 µg/mL inhibited CoQ biosynthesis without accumulation of apparent CoQ intermediates. This inhibition was counteracted by incubation with a 10-fold lower amount of PABA or PHB. Overexpression of PHB-polyprenyl transferase encoded by ppt1 (coq2) also overcame the inhibition of CoQ biosynthesis by Bz. Inhibition by Bz was efficient in S. pombe and Schizosaccharomyces japonicus, but less so in Saccharomyces cerevisiae, Aureobasidium pullulans, and Escherichia coli. Bz also inhibited a S. pombe ppt1 (coq2) deletion strain expressing human COQ2, and this strain also utilized PABA as a precursor of CoQ10. Thus, Bz is likely to inhibit prenylation reactions involving PHB or PABA catalyzed by Coq2.


Subject(s)
Benzoic Acid/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Fungal/drug effects , Schizosaccharomyces pombe Proteins/biosynthesis , Schizosaccharomyces/metabolism , Ubiquinone/biosynthesis , Prohibitins , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Ubiquinone/genetics
14.
Hum Mol Genet ; 29(19): 3296-3311, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32975579

ABSTRACT

Abnormalities of one carbon, glutathione and sulfide metabolisms have recently emerged as novel pathomechanisms in diseases with mitochondrial dysfunction. However, the mechanisms underlying these abnormalities are not clear. Also, we recently showed that sulfide oxidation is impaired in Coenzyme Q10 (CoQ10) deficiency. This finding leads us to hypothesize that the therapeutic effects of CoQ10, frequently administered to patients with primary or secondary mitochondrial dysfunction, might be due to its function as cofactor for sulfide:quinone oxidoreductase (SQOR), the first enzyme in the sulfide oxidation pathway. Here, using biased and unbiased approaches, we show that supraphysiological levels of CoQ10 induces an increase in the expression of SQOR in skin fibroblasts from control subjects and patients with mutations in Complex I subunits genes or CoQ biosynthetic genes. This increase of SQOR induces the downregulation of the cystathionine ß-synthase and cystathionine γ-lyase, two enzymes of the transsulfuration pathway, the subsequent downregulation of serine biosynthesis and the adaptation of other sulfide linked pathways, such as folate cycle, nucleotides metabolism and glutathione system. These metabolic changes are independent of the presence of sulfur aminoacids, are confirmed in mouse models, and are recapitulated by overexpression of SQOR, further proving that the metabolic effects of CoQ10 supplementation are mediated by the overexpression of SQOR. Our results contribute to a better understanding of how sulfide metabolism is integrated in one carbon metabolism and may explain some of the benefits of CoQ10 supplementation observed in mitochondrial diseases.


Subject(s)
Ataxia/pathology , Carbon/metabolism , Electron Transport Complex I/metabolism , Mitochondria/pathology , Mitochondrial Diseases/pathology , Muscle Weakness/pathology , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Sulfides/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Animals , Ataxia/genetics , Ataxia/metabolism , Electron Transport , Electron Transport Complex I/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Glutathione/metabolism , Humans , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Muscle Weakness/genetics , Muscle Weakness/metabolism , Oxidoreductases Acting on Sulfur Group Donors/genetics , Skin/drug effects , Skin/metabolism , Skin/pathology , Transcriptome , Ubiquinone/genetics , Ubiquinone/metabolism , Ubiquinone/pharmacology , Vitamins/pharmacology
15.
Molecules ; 25(13)2020 Jun 27.
Article in English | MEDLINE | ID: mdl-32605010

ABSTRACT

Coenzyme Q (CoQ) is an essential component of the mitochondrial electron transport chain and an important antioxidant present in all cellular membranes. CoQ deficiencies are frequent in aging and in age-related diseases, and current treatments are limited to CoQ supplementation. Strategies that rely on CoQ supplementation suffer from poor uptake and trafficking of this very hydrophobic molecule. In a previous study, the dietary flavonol kaempferol was reported to serve as a CoQ ring precursor and to increase the CoQ content in kidney cells, but neither the part of the molecule entering CoQ biosynthesis nor the mechanism were described. In this study, kaempferol labeled specifically in the B-ring was isolated from Arabidopsis plants. Kidney cells treated with this compound incorporated the B-ring of kaempferol into newly synthesized CoQ, suggesting that the B-ring is metabolized via a mechanism described in plant cells. Kaempferol is a natural flavonoid present in fruits and vegetables and possesses antioxidant, anticancer, and anti-inflammatory therapeutic properties. A better understanding of the role of kaempferol as a CoQ ring precursor makes this bioactive compound a potential candidate for the design of interventions aiming to increase endogenous CoQ biosynthesis and may improve CoQ deficient phenotypes in aging and disease.


Subject(s)
Antioxidants/metabolism , Ataxia/genetics , Kaempferols/metabolism , Mitochondrial Diseases/genetics , Muscle Weakness/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Animals , Ataxia/metabolism , Ataxia/pathology , Epithelial Cells/metabolism , Flavonols/metabolism , Humans , Kidney/metabolism , Kidney/pathology , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Membranes/metabolism , Muscle Weakness/metabolism , Muscle Weakness/pathology , Mutation/genetics , Ubiquinone/genetics , Ubiquinone/metabolism
16.
J Pediatr Endocrinol Metab ; 33(1): 165-170, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31821167

ABSTRACT

Background Coenzyme Q10 (CoQ10) serves as a shuttle for electrons from complexes I and II to complex III in the respiratory chain, and has important functions within the mitochondria. Primary CoQ10 deficiency is a mitochondrial disorder which has devastating effects, and which may be partially treated with exogenous CoQ10 supplementation. Case presentation A 9-month-old girl patient was referred to our clinic due to growth retardation, microcephaly and seizures. She was the third child of consanguineous parents (first-degree cousins) of Pakistani origin, born at 38 weeks gestation, weighing 2000 g after an uncomplicated pregnancy, and was hospitalized for 3 days due to respiratory distress. She had sustained clonic seizures when she was 4 months old. Physical examination showed microcephaly, truncal hypotonia and dysmorphic features. Metabolic tests were inconclusive. Abdominal ultrasonography revealed cystic appearance of the kidneys. Non-compaction of the left ventricle was detected in echocardiography. Cranial magnetic resonance imaging (MRI) showed hypoplasia of the cerebellar vermis and brain stem, corpus callosum agenesis, and cortical atrophy. A panel testing of 450 genes involved in inborn errors of metabolism (IEM) was performed that showed a novel frameshift c.384delG (Gly129Valfs*17) homozygous mutation in COQ9. A treatment of 5 mg/kg/day exogenous CoQ10 was started when she was 10 months old, and the dosage was increased to 50 mg/kg/day after the exact diagnosis. No objective neurological improvement could be observed after the adjustment of the drug dosage. Conclusions We report a case of CoQ10 deficiency due to a novel COQ9 gene mutation that adds clinical data from a newly diagnosed patient. Our case also outlines the importance of genetic panels used for specific diseases including IEM.


Subject(s)
Ataxia/etiology , Mitochondria/pathology , Mitochondrial Diseases/etiology , Muscle Weakness/etiology , Mutation , Rare Diseases/etiology , Ubiquinone/deficiency , Ataxia/pathology , Female , Humans , Infant , Mitochondria/genetics , Mitochondrial Diseases/pathology , Muscle Weakness/pathology , Prognosis , Rare Diseases/pathology , Ubiquinone/genetics
17.
Parkinsonism Relat Disord ; 68: 8-16, 2019 11.
Article in English | MEDLINE | ID: mdl-31621627

ABSTRACT

Cerebellar ataxia is a hallmark of coenzyme Q10 (CoQ10) deficiency associated with COQ8A mutations. We present four patients, one with novel COQ8A pathogenic variants all with early, prominent handwriting impairment, dystonia and only mild ataxia. To better define the phenotypic spectrum and course of COQ8A disease, we review the clinical presentation and evolution in 47 reported cases. Individuals with COQ8A mutation display great clinical variability and unpredictable responses to CoQ10 supplementation. Onset is typically during infancy or childhood with ataxic features associated with developmental delay or regression. When disease onset is later in life, first symptoms can include: incoordination, epilepsy, tremor, and deterioration of writing. The natural history is characterized by a progression to a multisystem brain disease dominated by ataxia, with disease severity inversely correlated with age at onset. Six previously reported cases share with ours, a clinical phenotype characterized by slowly progressive or static writing difficulties, focal dystonia, and speech disorder, with only minimal ataxia. The combination of writing difficulty, dystonia and ataxia is a distinctive constellation that is reminiscent of a previously described clinical entity called Dystonia Ataxia Syndrome (DYTCA) and is an important clinical indicator of COQ8A mutations, even when ataxia is mild or absent.


Subject(s)
Ataxia , Disease Progression , Dystonic Disorders , Handwriting , Heterozygote , Mitochondrial Diseases , Mitochondrial Proteins/genetics , Muscle Weakness , Ubiquinone/deficiency , Adult , Ataxia/complications , Ataxia/epidemiology , Ataxia/etiology , Ataxia/genetics , Ataxia/physiopathology , Child , Dystonic Disorders/epidemiology , Dystonic Disorders/etiology , Dystonic Disorders/genetics , Dystonic Disorders/physiopathology , Female , Humans , Male , Middle Aged , Mitochondrial Diseases/complications , Mitochondrial Diseases/epidemiology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/physiopathology , Muscle Weakness/complications , Muscle Weakness/epidemiology , Muscle Weakness/genetics , Muscle Weakness/physiopathology , Ubiquinone/genetics , Young Adult
18.
Oxid Med Cell Longev ; 2019: 3904905, 2019.
Article in English | MEDLINE | ID: mdl-31379988

ABSTRACT

Coenzyme Q (CoQ), a redox-active lipid, is comprised of a quinone group and a polyisoprenoid tail. It is an electron carrier in the mitochondrial respiratory chain, a cofactor of other mitochondrial dehydrogenases, and an essential antioxidant. CoQ requires a large set of enzymes for its biosynthesis; mutations in genes encoding these proteins cause primary CoQ deficiency, a clinically and genetically heterogeneous group of diseases. Patients with CoQ deficiency often respond to oral CoQ10 supplementation. Treatment is however problematic because of the low bioavailability of CoQ10 and the poor tissue delivery. In recent years, bypass therapy using analogues of the precursor of the aromatic ring of CoQ has been proposed as a promising alternative. We have previously shown using a yeast model that vanillic acid (VA) can bypass mutations of COQ6, a monooxygenase required for the hydroxylation of the C5 carbon of the ring. In this work, we have generated a human cell line lacking functional COQ6 using CRISPR/Cas9 technology. We show that these cells cannot synthesize CoQ and display severe ATP deficiency. Treatment with VA can recover CoQ biosynthesis and ATP production. Moreover, these cells display increased ROS production, which is only partially corrected by exogenous CoQ, while VA restores ROS to normal levels. Furthermore, we show that these cells accumulate 3-decaprenyl-1,4-benzoquinone, suggesting that in mammals, the decarboxylation and C1 hydroxylation reactions occur before or independently of the C5 hydroxylation. Finally, we show that COQ6 isoform c (transcript NM_182480) does not encode an active enzyme. VA can be produced in the liver by the oxidation of vanillin, a nontoxic compound commonly used as a food additive, and crosses the blood-brain barrier. These characteristics make it a promising compound for the treatment of patients with CoQ deficiency due to COQ6 mutations.


Subject(s)
Adenosine Triphosphate/metabolism , Ubiquinone/analogs & derivatives , Vanillic Acid/pharmacology , Amino Acid Sequence , Animals , CRISPR-Cas Systems/genetics , HEK293 Cells , Humans , Mitochondria/metabolism , Mutagenesis, Site-Directed , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Reactive Oxygen Species/metabolism , Sequence Alignment , Ubiquinone/biosynthesis , Ubiquinone/genetics , Ubiquinone/metabolism
19.
Sci Rep ; 9(1): 7995, 2019 05 29.
Article in English | MEDLINE | ID: mdl-31142763

ABSTRACT

We previously reported that oxidative stress induced by long-term tacrolimus treatment impairs mitochondrial function in pancreatic beta cells. In this study, we aimed to investigate the therapeutic potential of coenzyme Q10, which is known to be a powerful antioxidant, in mitochondrial dysfunction in tacrolimus-induced diabetic rats. In a rat model of tacrolimus-induced diabetes mellitus, coenzyme Q10 treatment improved pancreatic beta cell function. The administration of coenzyme Q10 improved insulin immunoreactivity within islets, which was accompanied by reductions in oxidative stress and apoptosis. Assessment of the mitochondrial ultrastructure by electron microscopy revealed that coenzyme Q10 treatment increased the size, number, and volume of mitochondria, as well as the number of insulin granules compared with that induced by tacrolimus treatment alone. An in vitro study using a pancreatic beta cell line showed that tacrolimus treatment increased apoptosis and the production of mitochondrial reactive oxygen species, while cotreatment with coenzyme Q10 effectively attenuated these alterations. At the subcellular level, tacrolimus-induced impairment of mitochondrial respiration was significantly improved by coenzyme Q10, as evidenced by the increased mitochondrial oxygen consumption and ATP production. Our data indicate that coenzyme Q10 plays an important role in reducing tacrolimus-induced oxidative stress and protects the mitochondria in pancreatic beta cells. These findings suggest that supplementation with coenzyme Q10 has beneficial effects in tacrolimus-induced diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Oxidative Stress/drug effects , Tacrolimus/adverse effects , Ubiquinone/analogs & derivatives , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Rats , Reactive Oxygen Species/metabolism , Ubiquinone/genetics , Ubiquinone/pharmacology
20.
J Lipid Res ; 60(7): 1293-1310, 2019 07.
Article in English | MEDLINE | ID: mdl-31048406

ABSTRACT

Coenzyme Q (CoQ or ubiquinone) serves as an essential redox-active lipid in respiratory electron and proton transport during cellular energy metabolism. CoQ also functions as a membrane-localized antioxidant protecting cells against lipid peroxidation. CoQ deficiency is associated with multiple human diseases; CoQ10 supplementation in particular has noted cardioprotective benefits. In Saccharomyces cerevisiae, Coq10, a putative START domain protein, is believed to chaperone CoQ to sites where it functions. Yeast coq10 deletion mutants (coq10Δ) synthesize CoQ inefficiently during log phase growth and are respiratory defective and sensitive to oxidative stress. Humans have two orthologs of yeast COQ10, COQ10A and COQ10B Here, we tested the human co-orthologs for their ability to rescue the yeast mutant. We showed that expression of either human ortholog, COQ10A or COQ10B, rescues yeast coq10Δ mutant phenotypes, restoring the function of respiratory-dependent growth on a nonfermentable carbon source and sensitivity to oxidative stress induced by treatment with PUFAs. These effects indicate a strong functional conservation of Coq10 across different organisms. However, neither COQ10A nor COQ10B restored CoQ biosynthesis when expressed in the yeast coq10Δ mutant. The involvement of yeast Coq10 in CoQ biosynthesis may rely on its interactions with another protein, possibly Coq11, which is not found in humans. Coexpression analyses of yeast COQ10 and human COQ10A and COQ10B provide additional insights to functions of these START domain proteins and their potential roles in other biologic pathways.


Subject(s)
Ataxia/metabolism , Mitochondrial Diseases/metabolism , Muscle Weakness/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Antioxidants/metabolism , Ataxia/genetics , Humans , Lipid Peroxidation/physiology , Mass Spectrometry , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Muscle Weakness/genetics , Oxidative Stress/genetics , Oxidative Stress/physiology , Phosphoproteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Ubiquinone/genetics , Ubiquinone/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL