1
|
Wang Y, Lilienfeldt N, Hekimi S. Understanding coenzyme Q. Physiol Rev 2024; 104:1533-1610. [PMID: 38722242 PMCID: PMC11495197 DOI: 10.1152/physrev.00040.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/08/2024] [Accepted: 05/01/2024] [Indexed: 08/11/2024] Open
Abstract
Coenzyme Q (CoQ), also known as ubiquinone, comprises a benzoquinone head group and a long isoprenoid side chain. It is thus extremely hydrophobic and resides in membranes. It is best known for its complex function as an electron transporter in the mitochondrial electron transport chain (ETC) but is also required for several other crucial cellular processes. In fact, CoQ appears to be central to the entire redox balance of the cell. Remarkably, its structure and therefore its properties have not changed from bacteria to vertebrates. In metazoans, it is synthesized in all cells and is found in most, and maybe all, biological membranes. CoQ is also known as a nutritional supplement, mostly because of its involvement with antioxidant defenses. However, whether there is any health benefit from oral consumption of CoQ is not well established. Here we review the function of CoQ as a redox-active molecule in the ETC and other enzymatic systems, its role as a prooxidant in reactive oxygen species generation, and its separate involvement in antioxidant mechanisms. We also review CoQ biosynthesis, which is particularly complex because of its extreme hydrophobicity, as well as the biological consequences of primary and secondary CoQ deficiency, including in human patients. Primary CoQ deficiency is a rare inborn condition due to mutation in CoQ biosynthetic genes. Secondary CoQ deficiency is much more common, as it accompanies a variety of pathological conditions, including mitochondrial disorders as well as aging. In this context, we discuss the importance, but also the great difficulty, of alleviating CoQ deficiency by CoQ supplementation.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | - Noah Lilienfeldt
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | - Siegfried Hekimi
- Department of Biology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
2
|
Pan P, Zhou N, Sun Y, Chen Z, Han J, Zhou W. The Spectrum of clinical manifestations in newborns with the COQ4 mutation: case series and literature review. Front Pediatr 2024; 12:1410133. [PMID: 39398416 PMCID: PMC11466766 DOI: 10.3389/fped.2024.1410133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Background Coenzyme Q10 (CoQ10) plays an important role in the electron transport chain within the human mitochondrial respiratory chain. The manifestations of this deficiency exhibit a diverse range. This study investigates the clinical manifestations of primary coenzyme Q10 deficiency in neonates with the COQ4 mutation to improve the diagnosis of the disease and the prognosis through targeted treatment. Methods We report 4 patients with primary coenzyme Q10 deficiency by COQ4 variants in neonates. A comprehensive literature search and review for original articles and case reports with COQ4 mutation published from January 1989 to November 2023 was performed through Pubmed. We review clinical manifestations, diagnostic approaches, and treatment monitoring in these and 20 previously reported patients. Results Within the cohort of four cases examined, three females and one male were identified from two distinct families. Specifically, case 1 and 2 consisted of monoamniotic twins. Cases 3 and 4 were siblings. A comprehensive review of 20 cases involving neonatal-onset COQ4 mutation was conducted. Half of the cases are Chinese. There was no statistically significant difference in the mortality between Chinese (9/12, 75%) and other regions (11/12, 91.7%) (P = 0.27). The survival time for the 24 cases was 60.0 ± 98.0 days (95% confidence interval CI: 0-252.0 days). The incidence of prenatal abnormalities in preterm infants was significantly higher than that in full-term infants (66.7% vs. 16.7%, P = 0.02). Hyperlactatemia was one of the most common manifestations, accounting for 75% of cases (18/24). Twenty of the 24 cases were diagnosed by whole exome sequencing. Only 9 patients received exogenous coenzyme Q10 treatment, and all the 4 surviving patients received coenzyme Q10 supplementation. Conclusion The prognosis of COQ4 mutation in the neonatal period indicates a low survival rate and an poor prognosis. This may be due to the incomplete understanding of the mechanism of how COQ4 gene defects lead to coenzyme Q10 deficiency and why CoQ10 supplementation does not respond well to treatment. To improve the diagnostic rate, in addition to genetic testing, mitochondrial functional verification should be prioritized in southern China, where the incidence is relatively high. It will facilitate more in-depth mechanistic studies.
Collapse
Affiliation(s)
- Pianpian Pan
- Nenoatal Intensive Care Unit, Guangzhou Wowen and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Na Zhou
- Heart Center, Guangzhou Wowen and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yi Sun
- Nenoatal Intensive Care Unit, Guangzhou Wowen and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhengrong Chen
- Pathology Department, Guangzhou Wowen and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jin Han
- Prenatal Diagnostic Center, Guangzhou Wowen and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei Zhou
- Nenoatal Intensive Care Unit, Guangzhou Wowen and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Corral-Sarasa J, Martínez-Gálvez JM, González-García P, Wendling O, Jiménez-Sánchez L, López-Herrador S, Quinzii CM, Díaz-Casado ME, López LC. 4-Hydroxybenzoic acid rescues multisystemic disease and perinatal lethality in a mouse model of mitochondrial disease. Cell Rep 2024; 43:114148. [PMID: 38697100 DOI: 10.1016/j.celrep.2024.114148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/03/2024] [Accepted: 04/09/2024] [Indexed: 05/04/2024] Open
Abstract
Coenzyme Q (CoQ) deficiency syndrome is conventionally treated with limited efficacy using exogenous CoQ10. Poor outcomes result from low absorption and bioavailability of CoQ10 and the clinical heterogenicity of the disease. Here, we demonstrate that supplementation with 4-hydroxybenzoic acid (4HB), the precursor of the benzoquinone ring in the CoQ biosynthetic pathway, completely rescues multisystemic disease and perinatal lethality in a mouse model of CoQ deficiency. 4HB stimulates endogenous CoQ biosynthesis in tissues of Coq2 mutant mice, normalizing mitochondrial function and rescuing cardiac insufficiency, edema, and neurodevelopmental delay. In contrast, exogenous CoQ10 supplementation falls short in fully restoring the phenotype. The treatment is translatable to human use, as proven by in vitro studies in skin fibroblasts from patients with pathogenic variants in COQ2. The therapeutic approach extends to other disorders characterized by deficiencies in the production of 4HB and early steps of CoQ biosynthesis and instances of secondary CoQ deficiency.
Collapse
Affiliation(s)
- Julia Corral-Sarasa
- Instituto de Investigación Biosanitaria ibs.Granada, 18016 Granada, Spain; Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain
| | - Juan Manuel Martínez-Gálvez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Biofisika Institute (CSIC, UBV-EHU) and Department of Biochemistry and Molecular Biology, University of Basque Country, 48940 Leioa, Spain
| | - Pilar González-García
- Instituto de Investigación Biosanitaria ibs.Granada, 18016 Granada, Spain; Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain
| | - Olivia Wendling
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), 1 rue Laurent Fries, 67404 Illkirch, France
| | | | - Sergio López-Herrador
- Instituto de Investigación Biosanitaria ibs.Granada, 18016 Granada, Spain; Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain
| | - Catarina M Quinzii
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - María Elena Díaz-Casado
- Instituto de Investigación Biosanitaria ibs.Granada, 18016 Granada, Spain; Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain
| | - Luis C López
- Instituto de Investigación Biosanitaria ibs.Granada, 18016 Granada, Spain; Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), 18016 Granada, Spain.
| |
Collapse
|
4
|
Hsu CJ, Lee WT. Epilepsy and Coenzyme Q10 deficiency with COQ4 variants. Epilepsy Behav 2023; 149:109498. [PMID: 37948995 DOI: 10.1016/j.yebeh.2023.109498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 10/14/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023]
Abstract
Coenzyme Q10 (CoQ10) is one of the essential substances for mitochondrial energy synthesis and extra-mitochondrial vital function. Primary CoQ10 deficiency is a rare disease resulting from interruption of CoQ10 biosynthetic pathway and biallelic COQ4 variants are one of the genetic etiologies recognized in this hereditary disorder. The clinical heterogenicity is broad with wide onset age from prenatal period to adulthood. The typical manifestations include early pharmacoresistant seizure, severe cognition and/or developmental delay, dystonia, ataxia, and spasticity. Patients may also have multisystemic involvements such as cardiomyopathy, lactic acidosis or gastro-esophageal regurgitation disease. Oral CoQ10 supplement is the major therapeutic medication currently. Among those patients, c.370G > A variant is the most common pathogenic variant detected, especially in Asian population. This phenomenon also suggests that this specific allele may be the founder variants in Asia. In this article, we report two siblings with infantile onset seizures, developmental delay, cardiomyopathy, and diffuse brain atrophy. Genetic analysis of both two cases revealed homozygous COQ4 c.370G > A (p.Gly124Ser) variants. We also review the clinical manifestations of primary CoQ10 deficiency patients and possible treatment categories, which are still under survey. As oral CoQ10 supplement may improve or stabilize disease severity, early precise diagnosis of primary CoQ10 deficiency and early treatment are the most important issues. This review article helps to further understand clinical spectrum and treatment categories of primary CoQ10 deficiency with COQ4 variant.
Collapse
Affiliation(s)
- Chia-Jui Hsu
- Department of Pediatrics, National Taiwan University Hsin-Chu Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Wang-Tso Lee
- Graduate Institute of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Pediatric Neurology, National Taiwan University Children's Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
5
|
Tippairote T, Bjørklund G, Gasmi A, Semenova Y, Peana M, Chirumbolo S, Hangan T. Combined Supplementation of Coenzyme Q 10 and Other Nutrients in Specific Medical Conditions. Nutrients 2022; 14:4383. [PMID: 36297067 PMCID: PMC9609170 DOI: 10.3390/nu14204383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/07/2022] [Accepted: 10/15/2022] [Indexed: 07/23/2023] Open
Abstract
Coenzyme Q10 (CoQ10) is a compound with a crucial role in mitochondrial bioenergetics and membrane antioxidant protection. Despite the ubiquitous endogenous biosynthesis, specific medical conditions are associated with low circulating CoQ10 levels. However, previous studies of oral CoQ10 supplementation yielded inconsistent outcomes. In this article, we reviewed previous CoQ10 trials, either single or in combination with other nutrients, and stratified the study participants according to their metabolic statuses and medical conditions. The CoQ10 supplementation trials in elders reported many favorable outcomes. However, the single intervention was less promising when the host metabolic statuses were worsening with the likelihood of multiple nutrient insufficiencies, as in patients with an established diagnosis of metabolic or immune-related disorders. On the contrary, the mixed CoQ10 supplementation with other interacting nutrients created more promising impacts in hosts with compromised nutrient reserves. Furthermore, the results of either single or combined intervention will be less promising in far-advanced conditions with established damage, such as neurodegenerative disorders or cancers. With the limited high-level evidence studies on each host metabolic category, we could only conclude that the considerations of whether to take supplementation varied by the individuals' metabolic status and their nutrient reserves. Further studies are warranted.
Collapse
Affiliation(s)
- Torsak Tippairote
- Department of Nutritional and Environmental Medicine, HP Medical Center, Bangkok 10540, Thailand
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Toften 24, 8610 Mo i Rana, Norway
| | - Amin Gasmi
- Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, 69100 Villeurbanne, France
| | - Yuliya Semenova
- School of Medicine, Nazarbayev University, Astana 020000, Kazakhstan
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, via Vienna 2, 07100 Sassari, Italy
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
- CONEM Scientific Secretary, Strada Le Grazie 9, 37134 Verona, Italy
| | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania
| |
Collapse
|
6
|
Wang Y, Hekimi S. The efficacy of coenzyme Q 10 treatment in alleviating the symptoms of primary coenzyme Q 10 deficiency: A systematic review. J Cell Mol Med 2022; 26:4635-4644. [PMID: 35985679 PMCID: PMC9443948 DOI: 10.1111/jcmm.17488] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/21/2022] [Accepted: 06/30/2022] [Indexed: 12/31/2022] Open
Abstract
Coenzyme Q10 (CoQ10 ) is necessary for mitochondrial electron transport. Mutations in CoQ10 biosynthetic genes cause primary CoQ10 deficiency (PCoQD) and manifest as mitochondrial disorders. It is often stated that PCoQD patients can be treated by oral CoQ10 supplementation. To test this, we compiled all studies describing PCoQD patients up to May 2022. We excluded studies with no data on CoQ10 treatment, or with insufficient description of effectiveness. Out of 303 PCoQD patients identified, we retained 89 cases, of which 24 reported improvements after CoQ10 treatment (27.0%). In five cases, the patient's condition was reported to deteriorate after halting of CoQ10 treatment. 12 cases reported improvement in the severity of ataxia and 5 cases in the severity of proteinuria. Only a subjective description of improvement was reported for 4 patients described as responding. All reported responses were partial improvements of only some symptoms. For PCoQD patients, CoQ10 supplementation is replacement therapy. Yet, there is only very weak evidence for the efficacy of the treatment. Our findings, thus, suggest a need for caution when seeking to justify the widespread use of CoQ10 for the treatment of any disease or as dietary supplement.
Collapse
Affiliation(s)
- Ying Wang
- Department of BiologyMcGill UniversityMontrealQuebecCanada
| | | |
Collapse
|
7
|
González-García P, Díaz-Casado ME, Hidalgo-Gutiérrez A, Jiménez-Sánchez L, Bakkali M, Barriocanal-Casado E, Escames G, Chiozzi RZ, Völlmy F, Zaal EA, Berkers CR, Heck AJR, López LC. The Q-junction and the inflammatory response are critical pathological and therapeutic factors in CoQ deficiency. Redox Biol 2022; 55:102403. [PMID: 35863266 PMCID: PMC9301574 DOI: 10.1016/j.redox.2022.102403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/07/2022] [Accepted: 07/10/2022] [Indexed: 11/24/2022] Open
Abstract
Defects in Coenzyme Q (CoQ) metabolism have been associated with primary mitochondrial disorders, neurodegenerative diseases and metabolic conditions. The consequences of CoQ deficiency have not been fully addressed, and effective treatment remains challenging. Here, we use mice with primary CoQ deficiency (Coq9R239X), and we demonstrate that CoQ deficiency profoundly alters the Q-junction, leading to extensive changes in the mitochondrial proteome and metabolism in the kidneys and, to a lesser extent, in the brain. CoQ deficiency also induces reactive gliosis, which mediates a neuroinflammatory response, both of which lead to an encephalopathic phenotype. Importantly, treatment with either vanillic acid (VA) or β-resorcylic acid (β-RA), two analogs of the natural precursor for CoQ biosynthesis, partially restores CoQ metabolism, particularly in the kidneys, and induces profound normalization of the mitochondrial proteome and metabolism, ultimately leading to reductions in gliosis, neuroinflammation and spongiosis and, consequently, reversing the phenotype. Together, these results provide key mechanistic insights into defects in CoQ metabolism and identify potential disease biomarkers. Furthermore, our findings clearly indicate that the use of analogs of the CoQ biosynthetic precursor is a promising alternative therapy for primary CoQ deficiency and has potential for use in the treatment of more common neurodegenerative and metabolic diseases that are associated with secondary CoQ deficiency.
Collapse
Affiliation(s)
- Pilar González-García
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016, Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016, Granada, Spain
| | - María Elena Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016, Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016, Granada, Spain
| | - Agustín Hidalgo-Gutiérrez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016, Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016, Granada, Spain
| | | | - Mohammed Bakkali
- Departamento de Genética, Facultad de Ciencias, Universidad de Granada, 18071, Granada, Spain
| | - Eliana Barriocanal-Casado
- GENYO, Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer-University of Granada-Andalusian Regional Government, 18016, Granada, Spain
| | - Germaine Escames
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016, Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016, Granada, Spain
| | - Riccardo Zenezini Chiozzi
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584CH, Utrecht, Netherlands; Netherlands Proteomics Centre, Padualaan 8, 3584 CH, Utrecht, the Netherlands
| | - Franziska Völlmy
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584CH, Utrecht, Netherlands; Netherlands Proteomics Centre, Padualaan 8, 3584 CH, Utrecht, the Netherlands
| | - Esther A Zaal
- Netherlands Proteomics Centre, Padualaan 8, 3584 CH, Utrecht, the Netherlands; Division of Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3508 TD, Utrecht, the Netherlands
| | - Celia R Berkers
- Netherlands Proteomics Centre, Padualaan 8, 3584 CH, Utrecht, the Netherlands; Division of Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3508 TD, Utrecht, the Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584CH, Utrecht, Netherlands; Netherlands Proteomics Centre, Padualaan 8, 3584 CH, Utrecht, the Netherlands
| | - Luis C López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016, Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016, Granada, Spain.
| |
Collapse
|
8
|
Plasma CoQ10 Status in Patients with Propionic Acidaemia and Possible Benefit of Treatment with Ubiquinol. Antioxidants (Basel) 2022; 11:antiox11081588. [PMID: 36009307 PMCID: PMC9405378 DOI: 10.3390/antiox11081588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/09/2022] [Accepted: 08/14/2022] [Indexed: 11/30/2022] Open
Abstract
Propionic acidaemia (PA) is an innate error of metabolism involving a deficiency in the enzyme propionyl-CoA carboxylase. Better control of acute decompensation episodes together with better treatment and monitoring have improved the prognosis of patients with this problem. However, long-term complications can arise in those in whom good metabolic control is achieved, the result of mitochondrial dysfunction caused by deficient anaplerosis, increased oxidative stress, and reduced antioxidative capacity. Coenzyme Q10 (CoQ10) is a nutritional supplement that has a notable antioxidative effect and has been shown to improve mitochondrial function. The present prospective, interventional study examines the plasma concentration of CoQ10 in patients with PA, their tolerance of such supplementation with ubiquinol, and its benefits. Seven patients with PA (aged 2.5 to 20 years, 4 males) received supplements of CoQ10 in the form of ubiquinol (10 mg/kg/day for 6 months). A total of 6/7 patients showed reduced plasma CoQ10 concentrations that normalized after supplementation with ubiquinol (p-value < 0.001), which was well tolerated. Urinary citrate levels markedly increased during the study (p-value: 0.001), together with elevation of citrate/methlycitrate ratio (p-value: 0.03). No other significant changes were seen in plasma or urine biomarkers of PA. PA patients showed a deficiency of plasma CoQ10, which supplementation with ubiquinol corrected. The urinary excretion of Krebs cycle intermediate citrate and the citrate/methylcitrate ratio significantly increased compared to the baseline, suggesting improvement in anaplerosis. This treatment was well tolerated and should be further investigated as a means of preventing the chronic complications associated with likely multifactorial mitochondrial dysfunction in PA.
Collapse
|
9
|
Kuriyama N, Nakamura T, Nakazawa H, Wen T, Berra L, Bittner EA, Goverman J, Kaneki M. Bioavailability of Reduced Coenzyme Q10 (Ubiquinol-10) in Burn Patients. Metabolites 2022; 12:metabo12070613. [PMID: 35888737 PMCID: PMC9321044 DOI: 10.3390/metabo12070613] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial dysfunction has been implicated in the pathogenesis of inflammation and multi-organ dysfunction in major trauma, including burn injury. Coenzyme Q10 (CoQ10) is a metabolite of the mevalonate pathway and an essential cofactor for the electron transport in the mitochondria. In addition, its reduced form (ubiquinol) functions as an antioxidant. Little is known as to whether oral CoQ10 supplementation effectively increases intracellular CoQ10 levels in humans. To study the bioavailability of CoQ10 supplementation, we conducted a randomized, double-blind, placebo-controlled study of reduced CoQ10 (ubiquinol-10) (1800 mg/day, t.i.d.) in burn patients at a single, tertiary-care hospital. Baseline plasma CoQ10 levels were significantly lower in burn patients than in healthy volunteers, although plasma CoQ10/cholesterol ratio did not differ between the groups. CoQ10 supplementation increased plasma concentrations of total and reduced CoQ10 and total CoQ10 content in peripheral blood mononuclear cells (PBMCs) in burn patients compared with the placebo group. CoQ10 supplementation did not significantly change circulating levels of mitochondrial DNA, inflammatory markers (e.g., interleukins, TNF-α, IFN-γ), or Sequential Organ Failure Assessment (SOFA) scores compared with the placebo group. This study showed that a relatively high dose of reduced CoQ10 supplementation increased the intracellular CoQ10 content in PBMCs as well as plasma concentrations in burn patients.
Collapse
Affiliation(s)
- Naohide Kuriyama
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA; (N.K.); (T.N.); (H.N.); (T.W.); (L.B.); (E.A.B.)
- Shriners Hospitals for Children, 51 Blossom Steet, Boston, MA 02114, USA
| | - Tomoyuki Nakamura
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA; (N.K.); (T.N.); (H.N.); (T.W.); (L.B.); (E.A.B.)
- Shriners Hospitals for Children, 51 Blossom Steet, Boston, MA 02114, USA
| | - Harumasa Nakazawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA; (N.K.); (T.N.); (H.N.); (T.W.); (L.B.); (E.A.B.)
- Shriners Hospitals for Children, 51 Blossom Steet, Boston, MA 02114, USA
| | - Tyler Wen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA; (N.K.); (T.N.); (H.N.); (T.W.); (L.B.); (E.A.B.)
- Vassar College, 124 Raymond Avenue, Poughkeepsie, NY 12604, USA
| | - Lorenzo Berra
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA; (N.K.); (T.N.); (H.N.); (T.W.); (L.B.); (E.A.B.)
| | - Edward A. Bittner
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA; (N.K.); (T.N.); (H.N.); (T.W.); (L.B.); (E.A.B.)
| | - Jeremy Goverman
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA;
| | - Masao Kaneki
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA; (N.K.); (T.N.); (H.N.); (T.W.); (L.B.); (E.A.B.)
- Shriners Hospitals for Children, 51 Blossom Steet, Boston, MA 02114, USA
- Correspondence: ; Tel.: +617-726-8122; Fax: 617-726-8134
| |
Collapse
|
10
|
Mechanisms and Therapeutic Effects of Benzoquinone Ring Analogs in Primary CoQ Deficiencies. Antioxidants (Basel) 2022; 11:antiox11040665. [PMID: 35453349 PMCID: PMC9029335 DOI: 10.3390/antiox11040665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 11/16/2022] Open
Abstract
Coenzyme Q (CoQ) is a conserved polyprenylated lipid composed of a redox-active benzoquinone ring and a long polyisoprenyl tail that serves as a membrane anchor. CoQ biosynthesis involves multiple steps, including multiple modifications of the precursor ring 4-hydroxybenzoic acid. Mutations in the enzymes involved in CoQ biosynthesis pathway result in primary coenzyme Q deficiencies, mitochondrial disorders whose clinical heterogenicity reflects the multiple biological function of CoQ. Patients with these disorders do not always respond to CoQ supplementation, and CoQ analogs have not been successful as alternative approaches. Progress made in understanding the CoQ biosynthesis pathway and studies of supplementation with 4-hydroxybenzoic acid ring analogs have opened a new area in the field of primary CoQ deficiencies treatment. Here, we will review these studies, focusing on efficacy of the different 4-hydroxybenzoic acid ring analogs, models in which they have been tested, and their mechanisms of action. Understanding how these compounds ameliorate biochemical, molecular, and/or clinical phenotypes of CoQ deficiencies is important to develop the most rational treatment for CoQ deficient patients, depending on their molecular defects.
Collapse
|
11
|
Xie J, Jiang J, Guo Q. Primary Coenzyme Q10 Deficiency-7 and Pathogenic COQ4 Variants: Clinical Presentation, Biochemical Analyses, and Treatment. Front Genet 2022; 12:776807. [PMID: 35154243 PMCID: PMC8826242 DOI: 10.3389/fgene.2021.776807] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
Primary Coenzyme Q10 Deficiency-7 (COQ10D7) is a rare mitochondrial disorder caused by pathogenic COQ4 variants. In this review, we discuss the correlation of COQ4 genotypes, particularly the East Asian-specific c.370G > A variant, with the clinical presentations and therapeutic effectiveness of coenzyme Q10 supplementation from an exon-dependent perspective. Pathogenic COQ4 variants in exons 1–4 are associated with less life-threating presentations, late onset, responsiveness to CoQ10 therapy, and a relatively long lifespan. In contrast, pathogenic COQ4 variants in exons 5–7 are associated with early onset, unresponsiveness to CoQ10 therapy, and early death and are more fatal. Patients with the East Asian-specific c.370G > A variant displays intermediate disease severity with multi-systemic dysfunction, which is between that of the patients with variants in exons 1–4 and 5–7. The mechanism underlying this exon-dependent genotype-phenotype correlation may be associated with the structure and function of COQ4. Sex is shown unlikely to be associated with disease severity. While point-of-care high-throughput sequencing would be useful for the rapid diagnosis of pathogenic COQ4 variants, whereas biochemical analyses of the characteristic impairments in CoQ10 biosynthesis and mitochondrial respiratory chain activity, as well as the phenotypic rescue of the CoQ10 treatment, are necessary to confirm the pathogenicity of suspicious variants. In addition to CoQ10 derivatives, targeted drugs and gene therapy could be useful treatments for COQ10D7 depending on the in-depth functional investigations and the development of gene editing technologies. This review provides a fundamental reference for the sub-classification of COQ10D7 and aim to advance our knowledge of the pathogenesis, clinical diagnosis, and prognosis of this disease and possible interventions.
Collapse
Affiliation(s)
- Jieqiong Xie
- United Diagnostic and Research Center for Clinical Genetics, Women and Children's Hospital, School of Medicine and School of Public Health, Xiamen University, Xiamen, China
| | - Jiayang Jiang
- United Diagnostic and Research Center for Clinical Genetics, Women and Children's Hospital, School of Medicine and School of Public Health, Xiamen University, Xiamen, China.,School of Medicine, Huaqiao University, Quanzhou, China
| | - Qiwei Guo
- United Diagnostic and Research Center for Clinical Genetics, Women and Children's Hospital, School of Medicine and School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
12
|
McCarty MF. Nutraceutical and Dietary Strategies for Up-Regulating Macroautophagy. Int J Mol Sci 2022; 23:2054. [PMID: 35216170 PMCID: PMC8875972 DOI: 10.3390/ijms23042054] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
Macroautophagy is a "cell cleansing" process that rids cells of protein aggregates and damaged organelles that may contribute to disease pathogenesis and the dysfunctions associated with aging. Measures which boost longevity and health span in rodents typically up-regulate macroautophagy, and it has often been suggested that safe strategies which can promote this process in humans may contribute to healthful aging. The kinase ULK1 serves as a trigger for autophagy initiation, and the transcription factors TFEB, FOXO1, ATF4 and CHOP promote expression of a number of proteins which mediate macroautophagy. Nutraceutical or dietary measures which stimulate AMPK, SIRT1, eIF5A, and that diminish the activities of AKT and mTORC1, can be expected to boost the activities of these pro-autophagic factors. The activity of AMPK can be stimulated with the phytochemical berberine. SIRT1 activation may be achieved with a range of agents, including ferulic acid, melatonin, urolithin A, N1-methylnicotinamide, nicotinamide riboside, and glucosamine; correction of ubiquinone deficiency may also be useful in this regard, as may dietary strategies such as time-restricted feeding or intermittent fasting. In the context of an age-related decrease in cellular polyamine levels, provision of exogenous spermidine can boost the hypusination reaction required for the appropriate post-translational modification of eIF5A. Low-protein plant-based diets could be expected to increase ATF4 and CHOP expression, while diminishing IGF-I-mediated activation of AKT and mTORC1. Hence, practical strategies for protecting health by up-regulating macroautophagy may be feasible.
Collapse
Affiliation(s)
- Mark F McCarty
- Catalytic Longevity Foundation, San Diego, CA 92109, USA
| |
Collapse
|
13
|
González-García P, Barriocanal-Casado E, Díaz-Casado ME, López-Herrador S, Hidalgo-Gutiérrez A, López LC. Animal Models of Coenzyme Q Deficiency: Mechanistic and Translational Learnings. Antioxidants (Basel) 2021; 10:antiox10111687. [PMID: 34829558 PMCID: PMC8614664 DOI: 10.3390/antiox10111687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/16/2022] Open
Abstract
Coenzyme Q (CoQ) is a vital lipophilic molecule that is endogenously synthesized in the mitochondria of each cell. The CoQ biosynthetic pathway is complex and not completely characterized, and it involves at least thirteen catalytic and regulatory proteins. Once it is synthesized, CoQ exerts a wide variety of mitochondrial and extramitochondrial functions thank to its redox capacity and its lipophilicity. Thus, low levels of CoQ cause diseases with heterogeneous clinical symptoms, which are not always understood. The decreased levels of CoQ may be primary caused by defects in the CoQ biosynthetic pathway or secondarily associated with other diseases. In both cases, the pathomechanisms are related to the CoQ functions, although further experimental evidence is required to establish this association. The conventional treatment for CoQ deficiencies is the high doses of oral CoQ10 supplementation, but this therapy is not effective for some specific clinical presentations, especially in those involving the nervous system. To better understand the CoQ biosynthetic pathway, the biological functions linked to CoQ and the pathomechanisms of CoQ deficiencies, and to improve the therapeutic outcomes of this syndrome, a variety of animal models have been generated and characterized in the last decade. In this review, we show all the animal models available, remarking on the most important outcomes that each model has provided. Finally, we also comment some gaps and future research directions related to CoQ metabolism and how the current and novel animal models may help in the development of future research studies.
Collapse
Affiliation(s)
- Pilar González-García
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
- Correspondence: (P.G.-G.); (L.C.L.)
| | - Eliana Barriocanal-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - María Elena Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Sergio López-Herrador
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Agustín Hidalgo-Gutiérrez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Luis C. López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
- Correspondence: (P.G.-G.); (L.C.L.)
| |
Collapse
|
14
|
β-RA Targets Mitochondrial Metabolism and Adipogenesis, Leading to Therapeutic Benefits against CoQ Deficiency and Age-Related Overweight. Biomedicines 2021; 9:biomedicines9101457. [PMID: 34680574 PMCID: PMC8533582 DOI: 10.3390/biomedicines9101457] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/01/2021] [Accepted: 10/09/2021] [Indexed: 11/17/2022] Open
Abstract
Primary mitochondrial diseases are caused by mutations in mitochondrial or nuclear genes, leading to the abnormal function of specific mitochondrial pathways. Mitochondrial dysfunction is also a secondary event in more common pathophysiological conditions, such as obesity and metabolic syndrome. In both cases, the improvement and management of mitochondrial homeostasis remain challenging. Here, we show that beta-resorcylic acid (β-RA), which is a natural phenolic compound, competed in vivo with 4-hydroxybenzoic acid, which is the natural precursor of coenzyme Q biosynthesis. This led to a decrease in demethoxyubiquinone, which is an intermediate metabolite of CoQ biosynthesis that is abnormally accumulated in Coq9R239X mice. As a consequence, β-RA rescued the phenotype of Coq9R239X mice, which is a model of primary mitochondrial encephalopathy. Moreover, we observed that long-term treatment with β-RA also reduced the size and content of the white adipose tissue (WAT) that is normally accumulated during aging in wild-type mice, leading to the prevention of hepatic steatosis and an increase in survival at the elderly stage of life. The reduction in WAT content was due to a decrease in adipogenesis, an adaptation of the mitochondrial proteome in the kidneys, and stimulation of glycolysis and acetyl-CoA metabolism. Therefore, our results demonstrate that β-RA acted through different cellular mechanisms, with effects on mitochondrial metabolism; as such, it may be used for the treatment of primary coenzyme Q deficiency, overweight, and hepatic steatosis.
Collapse
|
15
|
Kalenikova EI, Gorodetskaya EA, Obolenskaya ON, Shapavo NS, Makarov VG, Medvedev OS. Pharmacokinetics and Tissue Distribution of Oxidized and Reduced Coenzyme Q10 Upon Intravenous Administration. Pharm Chem J 2021. [DOI: 10.1007/s11094-021-02471-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
16
|
Cirilli I, Damiani E, Dludla PV, Hargreaves I, Marcheggiani F, Millichap LE, Orlando P, Silvestri S, Tiano L. Role of Coenzyme Q 10 in Health and Disease: An Update on the Last 10 Years (2010-2020). Antioxidants (Basel) 2021; 10:antiox10081325. [PMID: 34439573 PMCID: PMC8389239 DOI: 10.3390/antiox10081325] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
The present review focuses on preclinical and clinical studies conducted in the last decade that contribute to increasing knowledge on Coenzyme Q10's role in health and disease. Classical antioxidant and bioenergetic functions of the coenzyme have been taken into consideration, as well as novel mechanisms of action involving the redox-regulated activation of molecular pathways associated with anti-inflammatory activities. Cardiovascular research and fertility remain major fields of application of Coenzyme Q10, although novel applications, in particular in relation to topical application, are gaining considerable interest. In this respect, bioavailability represents a major challenge and the innovation in formulation aspects is gaining critical importance.
Collapse
Affiliation(s)
- Ilenia Cirilli
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy;
| | - Elisabetta Damiani
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (E.D.); (F.M.); (L.E.M.); (P.O.); (S.S.)
| | - Phiwayinkosi Vusi Dludla
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa;
| | - Iain Hargreaves
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK;
| | - Fabio Marcheggiani
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (E.D.); (F.M.); (L.E.M.); (P.O.); (S.S.)
| | - Lauren Elizabeth Millichap
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (E.D.); (F.M.); (L.E.M.); (P.O.); (S.S.)
| | - Patrick Orlando
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (E.D.); (F.M.); (L.E.M.); (P.O.); (S.S.)
| | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (E.D.); (F.M.); (L.E.M.); (P.O.); (S.S.)
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (E.D.); (F.M.); (L.E.M.); (P.O.); (S.S.)
- Correspondence: ; Tel.: +39-071-220-4394
| |
Collapse
|
17
|
Turton N, Bowers N, Khajeh S, Hargreaves IP, Heaton RA. Coenzyme Q10 and the exclusive club of diseases that show a limited response to treatment. Expert Opin Orphan Drugs 2021. [DOI: 10.1080/21678707.2021.1932459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Nadia Turton
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool UK
| | - Nathan Bowers
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool UK
| | - Sam Khajeh
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool UK
| | - Iain P Hargreaves
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool UK
| | - Robert A Heaton
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool UK
| |
Collapse
|
18
|
Hidalgo-Gutiérrez A, González-García P, Díaz-Casado ME, Barriocanal-Casado E, López-Herrador S, Quinzii CM, López LC. Metabolic Targets of Coenzyme Q10 in Mitochondria. Antioxidants (Basel) 2021; 10:520. [PMID: 33810539 PMCID: PMC8066821 DOI: 10.3390/antiox10040520] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/14/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Coenzyme Q10 (CoQ10) is classically viewed as an important endogenous antioxidant and key component of the mitochondrial respiratory chain. For this second function, CoQ molecules seem to be dynamically segmented in a pool attached and engulfed by the super-complexes I + III, and a free pool available for complex II or any other mitochondrial enzyme that uses CoQ as a cofactor. This CoQ-free pool is, therefore, used by enzymes that link the mitochondrial respiratory chain to other pathways, such as the pyrimidine de novo biosynthesis, fatty acid β-oxidation and amino acid catabolism, glycine metabolism, proline, glyoxylate and arginine metabolism, and sulfide oxidation metabolism. Some of these mitochondrial pathways are also connected to metabolic pathways in other compartments of the cell and, consequently, CoQ could indirectly modulate metabolic pathways located outside the mitochondria. Thus, we review the most relevant findings in all these metabolic functions of CoQ and their relations with the pathomechanisms of some metabolic diseases, highlighting some future perspectives and potential therapeutic implications.
Collapse
Affiliation(s)
- Agustín Hidalgo-Gutiérrez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Pilar González-García
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - María Elena Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Eliana Barriocanal-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Sergio López-Herrador
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Catarina M. Quinzii
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Luis C. López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| |
Collapse
|
19
|
Gueguen N, Baris O, Lenaers G, Reynier P, Spinazzi M. Secondary coenzyme Q deficiency in neurological disorders. Free Radic Biol Med 2021; 165:203-218. [PMID: 33450382 DOI: 10.1016/j.freeradbiomed.2021.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/31/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022]
Abstract
Coenzyme Q (CoQ) is a ubiquitous lipid serving essential cellular functions. It is the only component of the mitochondrial respiratory chain that can be exogenously absorbed. Here, we provide an overview of current knowledge, controversies, and open questions about CoQ intracellular and tissue distribution, in particular in brain and skeletal muscle. We discuss human neurological diseases and mouse models associated with secondary CoQ deficiency in these tissues and highlight pharmacokinetic and anatomical challenges in exogenous CoQ biodistribution, recent improvements in CoQ formulations and imaging, as well as alternative therapeutical strategies to CoQ supplementation. The last section proposes possible mechanisms underlying secondary CoQ deficiency in human diseases with emphasis on neurological and neuromuscular disorders.
Collapse
Affiliation(s)
- Naig Gueguen
- Unité Mixte de Recherche (UMR) MITOVASC, Centre National de la Recherche Scientifique (CNRS) 6015, Institut National de la Santé et de la Recherche Médicale (INSERM) U1083, University of Angers, 49933, Angers, France; Department of Biochemistry and Molecular Biology, CHU Angers, 49933, Angers, France
| | - Olivier Baris
- Unité Mixte de Recherche (UMR) MITOVASC, Centre National de la Recherche Scientifique (CNRS) 6015, Institut National de la Santé et de la Recherche Médicale (INSERM) U1083, University of Angers, 49933, Angers, France
| | - Guy Lenaers
- Unité Mixte de Recherche (UMR) MITOVASC, Centre National de la Recherche Scientifique (CNRS) 6015, Institut National de la Santé et de la Recherche Médicale (INSERM) U1083, University of Angers, 49933, Angers, France
| | - Pascal Reynier
- Unité Mixte de Recherche (UMR) MITOVASC, Centre National de la Recherche Scientifique (CNRS) 6015, Institut National de la Santé et de la Recherche Médicale (INSERM) U1083, University of Angers, 49933, Angers, France; Department of Biochemistry and Molecular Biology, CHU Angers, 49933, Angers, France
| | - Marco Spinazzi
- Unité Mixte de Recherche (UMR) MITOVASC, Centre National de la Recherche Scientifique (CNRS) 6015, Institut National de la Santé et de la Recherche Médicale (INSERM) U1083, University of Angers, 49933, Angers, France; Neuromuscular Reference Center, Department of Neurology, CHU Angers, 49933, Angers, France.
| |
Collapse
|
20
|
González-García P, Hidalgo-Gutiérrez A, Mascaraque C, Barriocanal-Casado E, Bakkali M, Ziosi M, Abdihankyzy UB, Sánchez-Hernández S, Escames G, Prokisch H, Martín F, Quinzii CM, López LC. Coenzyme Q10 modulates sulfide metabolism and links the mitochondrial respiratory chain to pathways associated to one carbon metabolism. Hum Mol Genet 2020; 29:3296-3311. [PMID: 32975579 PMCID: PMC7724311 DOI: 10.1093/hmg/ddaa214] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/03/2020] [Accepted: 09/23/2020] [Indexed: 01/14/2023] Open
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 β-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.
Collapse
Affiliation(s)
- Pilar González-García
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada 18016, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Agustín Hidalgo-Gutiérrez
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada 18016, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Cristina Mascaraque
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Eliana Barriocanal-Casado
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada 18016, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Mohammed Bakkali
- Departamento de Genética, Facultad de Ciencias, Universidad de Granada, Granada 18071, Spain
| | - Marcello Ziosi
- Department of Neurology, Columbia University Medical Center, New York 10032, NY, USA
| | | | | | - Germaine Escames
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada 18016, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Holger Prokisch
- Institute of Human Genetics, Technische Universität München, München 81675, Germany
| | - Francisco Martín
- Genomic Medicine Department, Centre for Genomics and Oncological Research, Granada 18007, Spain
| | - Catarina M Quinzii
- Department of Neurology, Columbia University Medical Center, New York 10032, NY, USA
| | - Luis C López
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada 18016, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| |
Collapse
|
21
|
Wang Y, Hekimi S. Micellization of coenzyme Q by the fungicide caspofungin allows for safe intravenous administration to reach extreme supraphysiological concentrations. Redox Biol 2020; 36:101680. [PMID: 32810741 PMCID: PMC7451649 DOI: 10.1016/j.redox.2020.101680] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
Coenzyme Q10 (CoQ10; also known as ubiquinone) is a vital, redox-active membrane component that functions as obligate electron transporter in the mitochondrial respiratory chain, as cofactor in other enzymatic processes and as antioxidant. CoQ10 supplementation has been widely investigated for treating a variety of acute and chronic conditions in which mitochondrial function or oxidative stress play a role. In addition, it is used as replacement therapy in patients with CoQ deficiency including inborn primary CoQ10 deficiency due to mutations in CoQ10-biosynthetic genes as well as secondary CoQ10 deficiency, which is frequently observed in patients with mitochondrial disease syndrome and in other conditions. However, despite many tests and some promising results, whether CoQ10 treatment is beneficial in any indication has remained inconclusive. Because CoQ10 is highly insoluble, it is only available in oral formulations, despite its very poor oral bioavailability. Using a novel model of CoQ-deficient cells, we screened a library of FDA-approved drugs for an activity that could increase the uptake of exogenous CoQ10 by the cell. We identified the fungicide caspofungin as capable of increasing the aqueous solubility of CoQ10 by several orders of magnitude. Caspofungin is a mild surfactant that solubilizes CoQ10 by forming nano-micelles with unique properties favoring stability and cellular uptake. Intravenous administration of the formulation in mice achieves unprecedented increases in CoQ10 plasma levels and in tissue uptake, with no observable toxicity. As it contains only two safe components (caspofungin and CoQ10), this injectable formulation presents a high potential for clinical safety and efficacy. Coenzyme Q10 (CoQ10) can be solubilized by the antifungal drug caspofungin (CF). CF is a mild surfactant and solubilizes CoQ10 in water by forming micellar structures with a high CoQ10 content. CF/CoQ10 micelles have unique properties favoring rapid and efficient uptake into cells and mitochondria. CF/CoQ10 micelles can be intravenously administrated without signs of toxicity. Intravenous administration of CF/CoQ10 in mice achieves unprecedented elevation of CoQ10 plasma levels and tissue uptake.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | - Siegfried Hekimi
- Department of Biology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
22
|
Kawashima C, Matsuzawa Y, Konishi M, Akiyama E, Suzuki H, Sato R, Nakahashi H, Kikuchi S, Kimura Y, Maejima N, Iwahashi N, Hibi K, Kosuge M, Ebina T, Tamura K, Kimura K. Ubiquinol Improves Endothelial Function in Patients with Heart Failure with Reduced Ejection Fraction: A Single-Center, Randomized Double-Blind Placebo-Controlled Crossover Pilot Study. Am J Cardiovasc Drugs 2020; 20:363-372. [PMID: 31713723 DOI: 10.1007/s40256-019-00384-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Endothelial dysfunction is reportedly associated with worse outcomes in patients with chronic heart failure. Ubiquinol is a reduced form of coenzyme Q10 (CoQ10) that may improve endothelial function. OBJECTIVE We assessed the hypothesis that ubiquinol improves peripheral endothelial function in patients with heart failure with reduced ejection fraction (HFrEF). METHODS In this randomized, double-blind, placebo-controlled, crossover pilot study, 14 patients with stable HFrEF were randomly and blindly allocated to ubiquinol 400 mg/day or placebo for 3 months. After a 1-month washout period, patients were crossed over to the alternative treatment. Before and after each treatment, we assessed peripheral endothelial function using the reactive hyperemia index (RHI) and analyzed it using the natural logarithm of RHI (LnRHI). RESULTS Peripheral endothelial function as assessed by LnRHI tended to improve with ubiquinol 400 mg/day for 3 months (p = 0.076). Original RHI values were also compared, and RHI significantly improved with ubiquinol treatment (pre-RHI 1.57 [interquartile range (IQR) 1.39-1.80], post-RHI 1.74 [IQR 1.63-2.02], p = 0.026), but not with placebo (pre-RHI 1.67 [IQR 1.53-1.85], post-RHI 1.51 [IQR 1.39-2.11], p = 0.198). CONCLUSIONS Ubiquinol 400 mg/day for 3 months led to significant improvement in peripheral endothelial function in patients with HFrEF. Ubiquinol may be a therapeutic option for individuals with HFrEF. Large-scale randomized controlled trials of CoQ10 supplementation in patients with HFrEF are needed. CLINICAL TRIAL REGISTRATION Japanese University Hospital Medical Information Network (UMIN-ICDR). Clinical Trial identifier number UMIN000012604.
Collapse
Affiliation(s)
- Chika Kawashima
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University School of Medicine, Yokohama, Japan
| | - Yasushi Matsuzawa
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan.
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University School of Medicine, Yokohama, Japan.
| | - Masaaki Konishi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Eiichi Akiyama
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Hiroyuki Suzuki
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Ryosuke Sato
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Hidefumi Nakahashi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Shinnosuke Kikuchi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Yuichiro Kimura
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Nobuhiko Maejima
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Noriaki Iwahashi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Masami Kosuge
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Toshiaki Ebina
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University School of Medicine, Yokohama, Japan
| | - Kazuo Kimura
- Division of Cardiology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan
| |
Collapse
|
23
|
Berardo A, Quinzii CM. Redefining infantile-onset multisystem phenotypes of coenzyme Q 10-deficiency in the next-generation sequencing era. JOURNAL OF TRANSLATIONAL GENETICS AND GENOMICS 2020; 4:22-35. [PMID: 33426503 PMCID: PMC7791541 DOI: 10.20517/jtgg.2020.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Primary coenzyme Q10 (CoQ10) deficiency encompasses a subset of mitochondrial diseases caused by mutations affecting proteins involved in the CoQ10 biosynthetic pathway. One of the most frequent clinical syndromes associated with primary CoQ10 deficiency is the severe infantile multisystemic form, which, until recently, was underdiagnosed. In the last few years, the availability of genetic screening through whole exome sequencing and whole genome sequencing has enabled molecular diagnosis in a growing number of patients with this syndrome and has revealed new disease phenotypes and molecular defects in CoQ10 biosynthetic pathway genes. Early genetic screening can rapidly and non-invasively diagnose primary CoQ10 deficiencies. Early diagnosis is particularly important in cases of CoQ10 deficient steroid-resistant nephrotic syndrome, which frequently improves with treatment. In contrast, the infantile multisystemic forms of CoQ10 deficiency, particularly when manifesting with encephalopathy, present therapeutic challenges, due to poor responses to CoQ10 supplementation. Administration of CoQ10 biosynthetic intermediate compounds is a promising alternative to CoQ10; however, further pre-clinical studies are needed to establish their safety and efficacy, as well as to elucidate the mechanism of actions of the intermediates. Here, we review the molecular defects causes of the multisystemic infantile phenotype of primary CoQ10 deficiency, genotype-phenotype correlations, and recent therapeutic advances.
Collapse
Affiliation(s)
- Andres Berardo
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Catarina M Quinzii
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
24
|
Abnormalities of hydrogen sulfide and glutathione pathways in mitochondrial dysfunction. J Adv Res 2020; 27:79-84. [PMID: 33318868 PMCID: PMC7728579 DOI: 10.1016/j.jare.2020.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/17/2022] Open
Abstract
Background Mitochondrial disorders are genetic diseases for which therapy remains woefully inadequate. Therapy of these disorders is particularly challenging partially due to the heterogeneity and tissue-specificity of pathomechanisms involved in these disorders. Abnormalities in hydrogen sulfide (H2S) metabolism are emerging as novel mechanism in mitochondrial dysfunction. However, further studies are necessary to understand the effects, protective or detrimental, of these abnormalities, and their relevance, in mitochondrial diseases. Aim of Review To review the recent evidences of derangement of the metabolism of H2S, at biosynthesis or oxidation levels, in mitochondrial dysfunction, focusing specifically on the alterations of H2S oxidation caused by primary Coenzyme Q (CoQ) deficiency. Key Scientific Concepts of Review Mitochondria play a key role in the regulation of H2S and GSH metabolism pathways. However, further studies are needed to understand the consequences of abnormalities of H2S and GSH synthesis on the oxidation pathway, and vice versa; and on the levels of H2S and GSH, their tissue-specific detrimental effects, and their role the role in mitochondrial diseases. Beside the known H2S pathways, additional, tissue-specific, enzymatic systems, involved in H2S production and elimination, might exist.
Collapse
|
25
|
Suzuki Y, Nagato S, Sakuraba K, Morio K, Sawaki K. Short-term ubiquinol-10 supplementation alleviates tissue damage in muscle and fatigue caused by strenuous exercise in male distance runners. INT J VITAM NUTR RES 2020; 91:261-270. [PMID: 32003645 DOI: 10.1024/0300-9831/a000627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Background: Coenzyme Q10 (CoQ10) is the electron transporter in oxidative phosphorylation and an endogenous antioxidant. Recent researches have indicated that doses of 200-300 mg/day are needed to recognize effects to prevent oxidative damage in athletes, and the reduced form of CoQ10, ubiquinol-10, is more bioavailable than its oxidized form. Therefore, we hypothesized that higher doses of ubiquinol-10 could elevate plasma CoQ10 levels rapidly and exert physiological benefits in athletes. Therefore, a placebo controlled, double blinded test was carried out to determine the effects of ubiquinol-10 on the extravasate enzymes and fatigue levels of distance runners. Methods: Sixteen male collegiate distance runners were allocated to two groups receiving 300 mg/day of ubiquinol-10 (19.8 ± 1.7 years) or a placebo (20.1 ± 1.6 years) for 12 days during summer training that comprised 25- and 40-km runs on days 7 and 9, respectively. Results: Ubiquinol-10 elevated plasma CoQ10 concentration to 5.62 μg/mL and significantly decreased activities of the serum extravasate enzymes, CK, ALT, LDH (P < 0.01), and AST (P < 0.05) on day 6. Subjective fatigue status was significantly elevated on day 10 (the day after the 45-km run) in the placebo group (P < 0.001), but did not significantly change in the group given ubiquinol-10. Therefore, ubiquinol-10 could mitigate tissue damage and alleviate fatigue status in distance runners during summer training. Conclusions: Ubiquinol-10 (300 mg/day) supplementation elevated plasma CoQ10 concentrations almost to plateau levels, decreased extravasate enzymes within six days, and suppressed the subjective fatigue in male distance runners.
Collapse
Affiliation(s)
- Yoshio Suzuki
- Juntendo University Graduate School of Health and Sports Science, Hiragagakuendai, Inzai, Chiba, Japan.,Juntendo University Faculty of Health and Sports Science, Hiragagakuendai, Inzai, Chiba, Japan
| | - Shunsuke Nagato
- Juntendo University Faculty of Health and Sports Science, Hiragagakuendai, Inzai, Chiba, Japan
| | - Keishoku Sakuraba
- Juntendo University Graduate School of Health and Sports Science, Hiragagakuendai, Inzai, Chiba, Japan.,Juntendo University Faculty of Health and Sports Science, Hiragagakuendai, Inzai, Chiba, Japan
| | - Katsuya Morio
- Juntendo University Faculty of Health and Sports Science, Hiragagakuendai, Inzai, Chiba, Japan
| | - Keisuke Sawaki
- Juntendo University Faculty of Health and Sports Science, Hiragagakuendai, Inzai, Chiba, Japan
| |
Collapse
|
26
|
Niu YJ, Zhou W, Nie ZW, Zhou D, Xu YN, Ock SA, Yan CG, Cui XS. Ubiquinol-10 delays postovulatory oocyte aging by improving mitochondrial renewal in pigs. Aging (Albany NY) 2020; 12:1256-1271. [PMID: 31958774 PMCID: PMC7053629 DOI: 10.18632/aging.102681] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 12/25/2019] [Indexed: 02/02/2023]
Abstract
Ubiquinol-10, the reduced form of coenzyme Q10, protects mammalian cells from oxidative damage and enhances mitochondrial activity. However, the protective effect of ubiquinol-10 on mammalian oocytes is not well understood. In this study, we investigated the effect of ubiquinol-10 on porcine oocytes during postovulatory aging. Metaphase II oocytes were selected as fresh oocytes and further cultured for 48 h with different concentrations of ubiquinol-10 (0–400 μM) in vitro as a postovulatory aging model. After choosing the optimal concentration of ubiquinol-10 (100 μM) that maintained oocyte morphology and developmental competence during the progression of aging, the oocytes were randomly divided into five groups: fresh, control-24 h, ubiquinol-24 h, control-48 h, and ubiquinol-48 h. The results revealed that ubiquinol-10 significantly prevented aging-induced oxidative stress, GSH reduction, cytoskeleton impairment, apoptosis, and autophagy. Mitochondrial biogenesis (SIRT1 and PGC-1α) and mitophagy (PINK1 and PARKIN)-related proteins were decreased during aging. Addition of ubiquinol-10 prevented the aging-induced reduction of these proteins. Consequently, although mitochondrial content was decreased, the number of active mitochondria and ATP level were significantly increased upon treatment with ubiquinol-10. Thus, ubiquinol-10 has beneficial effects on porcine postovulatory aging oocytes owing to its antioxidant properties and ability to promote mitochondrial renewal.
Collapse
Affiliation(s)
- Ying-Jie Niu
- Department of Animal Science, Chungbuk National University, Cheongju, South Korea
| | - Wenjun Zhou
- Department of Animal Science, Chungbuk National University, Cheongju, South Korea
| | - Zheng-Wen Nie
- Department of Animal Science, Chungbuk National University, Cheongju, South Korea
| | - Dongjie Zhou
- Department of Animal Science, Chungbuk National University, Cheongju, South Korea
| | - Yong-Nan Xu
- College of Agriculture, Yanbian University, Yanji, China
| | - Sun A Ock
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeonju, South Korea
| | - Chang-Guo Yan
- College of Agriculture, Yanbian University, Yanji, China
| | - Xiang-Shun Cui
- Department of Animal Science, Chungbuk National University, Cheongju, South Korea
| |
Collapse
|
27
|
Díaz-Casado ME, Quiles JL, Barriocanal-Casado E, González-García P, Battino M, López LC, Varela-López A. The Paradox of Coenzyme Q 10 in Aging. Nutrients 2019; 11:nu11092221. [PMID: 31540029 PMCID: PMC6770889 DOI: 10.3390/nu11092221] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/06/2019] [Accepted: 09/08/2019] [Indexed: 12/14/2022] Open
Abstract
Coenzyme Q (CoQ) is an essential endogenously synthesized molecule that links different metabolic pathways to mitochondrial energy production thanks to its location in the mitochondrial inner membrane and its redox capacity, which also provide it with the capability to work as an antioxidant. Although defects in CoQ biosynthesis in human and mouse models cause CoQ deficiency syndrome, some animals models with particular defects in the CoQ biosynthetic pathway have shown an increase in life span, a fact that has been attributed to the concept of mitohormesis. Paradoxically, CoQ levels decline in some tissues in human and rodents during aging and coenzyme Q10 (CoQ10) supplementation has shown benefits as an anti-aging agent, especially under certain conditions associated with increased oxidative stress. Also, CoQ10 has shown therapeutic benefits in aging-related disorders, particularly in cardiovascular and metabolic diseases. Thus, we discuss the paradox of health benefits due to a defect in the CoQ biosynthetic pathway or exogenous supplementation of CoQ10.
Collapse
Affiliation(s)
- M Elena Díaz-Casado
- Institute of Biotechnology, Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 18016 Granada, Spain.
| | - José L Quiles
- Institute of Nutrition and Food Technology "José Mataix Verdú", Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
| | - Eliana Barriocanal-Casado
- Institute of Biotechnology, Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 18016 Granada, Spain.
| | - Pilar González-García
- Institute of Biotechnology, Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 18016 Granada, Spain.
| | - Maurizio Battino
- Department of Clinical Sicences, Università Politecnica delle Marche, 60131 Ancona, Italy.
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain.
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China.
| | - Luis C López
- Institute of Biotechnology, Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 18016 Granada, Spain.
| | - Alfonso Varela-López
- Institute of Nutrition and Food Technology "José Mataix Verdú", Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
| |
Collapse
|
28
|
Heaton R, Millichap L, Saleem F, Gannon J, Begum G, Hargreaves IP. Current biochemical treatments of mitochondrial respiratory chain disorders. Expert Opin Orphan Drugs 2019. [DOI: 10.1080/21678707.2019.1638250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Robert Heaton
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Lauren Millichap
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Fatima Saleem
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Jennifer Gannon
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Gemma Begum
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Iain P. Hargreaves
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
29
|
Cerqua C, Casarin A, Pierrel F, Vazquez Fonseca L, Viola G, Salviati L, Trevisson E. Vitamin K2 cannot substitute Coenzyme Q 10 as electron carrier in the mitochondrial respiratory chain of mammalian cells. Sci Rep 2019; 9:6553. [PMID: 31024065 PMCID: PMC6484000 DOI: 10.1038/s41598-019-43014-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/11/2019] [Indexed: 12/16/2022] Open
Abstract
Coenzyme Q10 (CoQ10) deficiencies are a group of heterogeneous conditions that respond to ubiquinone administration if treated soon after the onset of symptoms. However, this treatment is only partially effective due to its poor bioavailability. We tested whether vitamin K2, which was reported to act as a mitochondrial electron carrier in D. melanogaster, could mimic ubiquinone function in human CoQ10 deficient cell lines, and in yeast carrying mutations in genes required for coenzyme Q6 (CoQ6) biosynthesis. We found that vitamin K2, despite entering into mitochondria, restored neither electron flow in the respiratory chain, nor ATP synthesis. Conversely, coenzyme Q4 (CoQ4), an analog of CoQ10 with a shorter isoprenoid side chain, could efficiently substitute its function. Given its better solubility, CoQ4 could represent an alternative to CoQ10 in patients with both primary and secondary CoQ10 deficiencies.
Collapse
Affiliation(s)
- Cristina Cerqua
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy.,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy
| | - Alberto Casarin
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy.,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy
| | - Fabien Pierrel
- Univ. Grenoble Alpes, CNRS, CHU Grenoble Alpes, Grenoble INP, TIMC-IMAG, 38000, Grenoble, France
| | - Luis Vazquez Fonseca
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy.,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy
| | - Giampiero Viola
- Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.,Pediatric Hematooncology Laboratory, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy
| | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy. .,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.
| | - Eva Trevisson
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy. .,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.
| |
Collapse
|
30
|
Safety Assessment of Ubiquinol Acetate: Subchronic Toxicity and Genotoxicity Studies. J Toxicol 2019; 2019:3680757. [PMID: 31057608 PMCID: PMC6463567 DOI: 10.1155/2019/3680757] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 02/06/2019] [Indexed: 01/30/2023] Open
Abstract
Coenzyme Q10 (CoQ10) is a lipid soluble, endogenous antioxidant present at highest levels in the heart followed by the kidney and liver. The reduced CoQ10 ubiquinol is well known for its chemical instability and low bioavailability. The present study was designed to synthesize ubiquinol acetate, which is more stable and biologically active, and further evaluate its safety and genotoxic potential. Synthesized ubiquinol acetate showed better stability than that of ubiquinol at the end of 3 months. In vitro genotoxicity studies (AMES test, in vitro micronucleus and chromosomal aberration) showed ubiquinol acetate as nongenotoxic with no clastogenic or aneugenic effects at high dose of 5000 and 62.5 μg/mL, respectively. In subchronic toxicity study, ubiquinol acetate was administered orally to Sprague Dawley rats at 150, 300, and 600 mg/kg/day for 90 days. No treatment related adverse effects were observed in males at 600 mg/kg/day; however, females showed treatment related increase in AST and ALT with small focal irregular white-yellow spots in liver on gross necropsy examination. Histopathological evaluation revealed hepatocellular necrosis in high dose females which was considered as adverse. Based on the results, the No-Observed-Adverse-Effect Level (NOAEL) of ubiquinol acetate in males and females was determined as 600 and 300 mg/kg/day, respectively.
Collapse
|
31
|
Barriocanal-Casado E, Hidalgo-Gutiérrez A, Raimundo N, González-García P, Acuña-Castroviejo D, Escames G, López LC. Rapamycin administration is not a valid therapeutic strategy for every case of mitochondrial disease. EBioMedicine 2019; 42:511-523. [PMID: 30898651 PMCID: PMC6492073 DOI: 10.1016/j.ebiom.2019.03.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/09/2019] [Accepted: 03/11/2019] [Indexed: 12/30/2022] Open
Abstract
Background The vast majority of mitochondrial disorders have limited the clinical management to palliative care. Rapamycin has emerged as a potential therapeutic drug for mitochondrial diseases since it has shown therapeutic benefits in a few mouse models of mitochondrial disorders. However, the underlying therapeutic mechanism is unclear, the minimal effective dose needs to be defined and whether this therapy can be generally used is unknown. Methods We have evaluated whether low and high doses of rapamycin administration may result in therapeutic effects in a mouse model (Coq9R239X) of mitochondrial encephalopathy due to CoQ deficiency. The evaluation involved phenotypic, molecular, image (histopathology and MRI), metabolomics, transcriptomics and bioenergetics analyses. Findings Low dose of rapamycin induces metabolic changes in liver and transcriptomics modifications in midbrain. The high dose of rapamycin induces further changes in the transcriptomics profile in midbrain due to the general inhibition of mTORC1. However, neither low nor high dose of rapamycin were able to improve the mitochondrial bioenergetics, the brain injuries and the phenotypic characteristics of Coq9R239X mice, resulting in the lack of efficacy for increasing the survival. Interpretation These results may be due to the lack of microgliosis-derived neuroinflammation, the limitation to induce autophagy, or the need of a functional CoQ-junction. Therefore, the translation of rapamycin therapy into the clinic for patients with mitochondrial disorders requires, at least, the consideration of the particularities of each mitochondrial disease. Fund Supported by the grants from “Fundación Isabel Gemio - Federación Española de Enfermedades Neuromusculares – Federación FEDER” (TSR-1), the NIH (P01HD080642) and the ERC (Stg-337327).
Collapse
Affiliation(s)
- Eliana Barriocanal-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain
| | - Agustín Hidalgo-Gutiérrez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain
| | - Nuno Raimundo
- Universitätsmedizin Göttingen, Institute fur Zellbiochemie, Humboldtallee 23, room 01.423, 37073 Göttingen, Germany
| | - Pilar González-García
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain
| | - Darío Acuña-Castroviejo
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Spain
| | - Germaine Escames
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Spain
| | - Luis C López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Spain.
| |
Collapse
|
32
|
Water-soluble CoQ10 as A Promising Anti-aging Agent for Neurological Dysfunction in Brain Mitochondria. Antioxidants (Basel) 2019; 8:antiox8030061. [PMID: 30862106 PMCID: PMC6466529 DOI: 10.3390/antiox8030061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/19/2019] [Accepted: 03/08/2019] [Indexed: 11/17/2022] Open
Abstract
Mitochondrial function has been closely associated with normal aging and age-related diseases. Age-associated declines in mitochondrial function, such as changes in oxygen consumption rate, cytochrome c oxidase activity of complex IV, and mitochondrial coenzyme Q (CoQ) levels, begin as early as 12 to 15 months of age in male mouse brains. Brain mitochondrial dysfunction is accompanied by increased accumulation of phosphorylated α-synuclein in the motor cortex and impairment of motor activities, which are similar characteristics of Parkinson's disease. However, these age-associated defects are completely rescued by the administration of exogenous CoQ10 to middle-aged mice via its water solubilization by emulsification in drinking water. Further efforts to develop strategies to enhance the biological availability of CoQ10 to successfully ameliorate age-related brain mitochondrial dysfunction or neurodegenerative disorders may provide a promising anti-aging agent.
Collapse
|
33
|
Hidalgo-Gutiérrez A, Barriocanal-Casado E, Bakkali M, Díaz-Casado ME, Sánchez-Maldonado L, Romero M, Sayed RK, Prehn C, Escames G, Duarte J, Acuña-Castroviejo D, López LC. β-RA reduces DMQ/CoQ ratio and rescues the encephalopathic phenotype in Coq9R239X mice. EMBO Mol Med 2019; 11:e9466. [PMID: 30482867 PMCID: PMC6328940 DOI: 10.15252/emmm.201809466] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 10/23/2018] [Accepted: 10/26/2018] [Indexed: 01/15/2023] Open
Abstract
Coenzyme Q (CoQ) deficiency has been associated with primary defects in the CoQ biosynthetic pathway or to secondary events. In some cases, the exogenous CoQ supplementation has limited efficacy. In the Coq9R239X mouse model with fatal mitochondrial encephalopathy due to CoQ deficiency, we have tested the therapeutic potential of β-resorcylic acid (β-RA), a structural analog of the CoQ precursor 4-hydroxybenzoic acid and the anti-inflammatory salicylic acid. β-RA noticeably rescued the phenotypic, morphological, and histopathological signs of the encephalopathy, leading to a significant increase in the survival. Those effects were due to the decrease of the levels of demethoxyubiquinone-9 (DMQ9) and the increase of mitochondrial bioenergetics in peripheral tissues. However, neither CoQ biosynthesis nor mitochondrial function changed in the brain after the therapy, suggesting that some endocrine interactions may induce the reduction of the astrogliosis, spongiosis, and the secondary down-regulation of astrocytes-related neuroinflammatory genes. Because the therapeutic outcomes of β-RA administration were superior to those after CoQ10 supplementation, its use in the clinic should be considered in CoQ deficiencies.
Collapse
Affiliation(s)
- Agustín Hidalgo-Gutiérrez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Eliana Barriocanal-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Mohammed Bakkali
- Departamento de Genética, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - M Elena Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Laura Sánchez-Maldonado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Miguel Romero
- Departamento de Farmacología, Facultad de Farmacia, Universidad de Granada, Granada, Spain
| | - Ramy K Sayed
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Cornelia Prehn
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Germaine Escames
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Granada, Spain
| | - Juan Duarte
- Departamento de Farmacología, Facultad de Farmacia, Universidad de Granada, Granada, Spain
| | - Darío Acuña-Castroviejo
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Granada, Spain
| | - Luis C López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Granada, Spain
| |
Collapse
|
34
|
Barca E, Emmanuele V, DiMauro S, Toscano A, Quinzii CM. Anti-Oxidant Drugs: Novelties and Clinical Implications in Cerebellar Ataxias. Curr Neuropharmacol 2019; 17:21-32. [PMID: 29119930 PMCID: PMC6341493 DOI: 10.2174/1570159x15666171109125643] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 01/01/1970] [Accepted: 11/07/2017] [Indexed: 12/06/2022] Open
Abstract
BACKGROUND Hereditary cerebellar ataxias are a group of disorders characterized by heterogeneous clinical manifestations, progressive clinical course, and diverse genetic causes. No disease modifying treatments are yet available for many of these disorders. Oxidative stress has been recurrently identified in different progressive cerebellar diseases, and it represents a widely investigated target for treatment. OBJECTIVE To review the main aspects and new perspectives of antioxidant therapy in cerebellar ataxias ranging from bench to bedside. METHOD This article is a summary of the state-of-the-art on the use of antioxidant molecules in cerebellar ataxia treatments. It also briefly summarizes aspects of oxidative stress production and general characteristics of antioxidant compounds. RESULTS Antioxidants represent a vast category of compounds; old drugs have been extensively studied and modified in order to achieve better biological effects. Despite the vast body of literature present on the use of antioxidants in cerebellar ataxias, for the majority of these disorders conclusive results on the efficacy are still missing. CONCLUSION Antioxidant therapy in cerebellar ataxias is a promising field of investigations. To achieve the success in identifying the correct treatment more work needs to be done. In particular, a combined effort is needed by basic scientists in developing more efficient molecules, and by clinical researchers together with patients communities, to run clinical trials in order to identify conclusive treatments strategies.
Collapse
Affiliation(s)
- Emanuele Barca
- Address correspondence to this author at the Department of Neurology, Columbia University Medical Center, 630 W 168 Street, P&S 4-424/A, New York, NY 10032, USA; Tel: +1-212-305-1637; Fax: +1-212-305-3986; E-mail:
| | | | | | | | | |
Collapse
|
35
|
Reduction in the levels of CoQ biosynthetic proteins is related to an increase in lifespan without evidence of hepatic mitohormesis. Sci Rep 2018; 8:14013. [PMID: 30228311 PMCID: PMC6143522 DOI: 10.1038/s41598-018-32190-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/26/2018] [Indexed: 01/05/2023] Open
Abstract
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.
Collapse
|
36
|
Kleiner G, Barca E, Ziosi M, Emmanuele V, Xu Y, Hidalgo-Gutierrez A, Qiao C, Tadesse S, Area-Gomez E, Lopez LC, Quinzii CM. CoQ 10 supplementation rescues nephrotic syndrome through normalization of H 2S oxidation pathway. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3708-3722. [PMID: 30251690 DOI: 10.1016/j.bbadis.2018.09.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/03/2018] [Accepted: 09/05/2018] [Indexed: 12/11/2022]
Abstract
Nephrotic syndrome (NS), a frequent chronic kidney disease in children and young adults, is the most common phenotype associated with primary coenzyme Q10 (CoQ10) deficiency and is very responsive to CoQ10 supplementation, although the pathomechanism is not clear. Here, using a mouse model of CoQ deficiency-associated NS, we show that long-term oral CoQ10 supplementation prevents kidney failure by rescuing defects of sulfides oxidation and ameliorating oxidative stress, despite only incomplete normalization of kidney CoQ levels and lack of rescue of CoQ-dependent respiratory enzymes activities. Liver and kidney lipidomics, and urine metabolomics analyses, did not show CoQ metabolites. To further demonstrate that sulfides metabolism defects cause oxidative stress in CoQ deficiency, we show that silencing of sulfide quinone oxido-reductase (SQOR) in wild-type HeLa cells leads to similar increases of reactive oxygen species (ROS) observed in HeLa cells depleted of the CoQ biosynthesis regulatory protein COQ8A. While CoQ10 supplementation of COQ8A depleted cells decreases ROS and increases SQOR protein levels, knock-down of SQOR prevents CoQ10 antioxidant effects. We conclude that kidney failure in CoQ deficiency-associated NS is caused by oxidative stress mediated by impaired sulfides oxidation and propose that CoQ supplementation does not significantly increase the kidney pool of CoQ bound to the respiratory supercomplexes, but rather enhances the free pool of CoQ, which stabilizes SQOR protein levels rescuing oxidative stress.
Collapse
Affiliation(s)
- Giulio Kleiner
- Department of Neurology, Columbia University Medical Center, New York, NY, United States
| | - Emanuele Barca
- Department of Neurology, Columbia University Medical Center, New York, NY, United States
| | - Marcello Ziosi
- Department of Neurology, Columbia University Medical Center, New York, NY, United States
| | - Valentina Emmanuele
- Department of Neurology, Columbia University Medical Center, New York, NY, United States
| | - Yimeng Xu
- Department of Pathology, Columbia University Medical Center, New York, NY, United States
| | | | - Changhong Qiao
- Irving Institute for Clinical and Translational Research, Columbia University Medical Center, New York, NY, United States
| | - Saba Tadesse
- Department of Neurology, Columbia University Medical Center, New York, NY, United States
| | - Estela Area-Gomez
- Department of Neurology, Columbia University Medical Center, New York, NY, United States
| | - Luis C Lopez
- Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Catarina M Quinzii
- Department of Neurology, Columbia University Medical Center, New York, NY, United States.
| |
Collapse
|
37
|
López-Lluch G, Del Pozo-Cruz J, Sánchez-Cuesta A, Cortés-Rodríguez AB, Navas P. Bioavailability of coenzyme Q10 supplements depends on carrier lipids and solubilization. Nutrition 2018; 57:133-140. [PMID: 30153575 DOI: 10.1016/j.nut.2018.05.020] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/17/2018] [Accepted: 05/22/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Bioavailability of supplements with coenzyme Q10 (CoQ10) in humans seems to depend on the excipients of formulations and on physiological characteristics of the individuals. The aim of this study was to determine which factors presented in CoQ10 supplements affect the different response to CoQ10 in humans. METHODS We tested seven different supplement formulations containing 100 mg of CoQ10 in 14 young, healthy individuals. Bioavailability was measured as area under the curve of plasma CoQ10 levels over 48 h after ingestion of a single dose. Measurements were repeated in the same group of 14 volunteers in a double-blind crossover design with a minimum of 4 wk washout between intakes. RESULTS Bioavailability of the formulations showed large differences that were statistically significant. The two best absorbable formulations were soft-gel capsules containing ubiquinone (oxidized CoQ10) or ubiquinol (reduced CoQ10). The matrix used to dissolve CoQ10 and the proportion and addition of preservatives such as vitamin C affected the bioavailability of CoQ10. Although control measurements documented that all formulations contained 100 mg of either CoQ10 or ubiquinol, some of the participants showed high and others lower capacity to reach high increase of CoQ10 in blood, indicating the participation of individual unknown physiological factors. CONCLUSION This study highlights the importance of individually adapted selection of best formulations to reach the highest bioavailability of CoQ10 in humans.
Collapse
Affiliation(s)
- Guillermo López-Lluch
- Universidad Pablo de Olavide, Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Sevilla, Spain.
| | | | - Ana Sánchez-Cuesta
- Universidad Pablo de Olavide, Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Ana Belén Cortés-Rodríguez
- Universidad Pablo de Olavide, Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Plácido Navas
- Universidad Pablo de Olavide, Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| |
Collapse
|
38
|
Preclinical Study of the Pharmacokinetics of a New Intravenous Dosage Form of Ubiquinol. Pharm Chem J 2018. [DOI: 10.1007/s11094-018-1721-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
39
|
Perlman S, Boltshauser E. Drug treatment. ACTA ACUST UNITED AC 2018; 155:371-377. [DOI: 10.1016/b978-0-444-64189-2.00024-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
40
|
Luna-Sánchez M, Hidalgo-Gutiérrez A, Hildebrandt TM, Chaves-Serrano J, Barriocanal-Casado E, Santos-Fandila Á, Romero M, Sayed RK, Duarte J, Prokisch H, Schuelke M, Distelmaier F, Escames G, Acuña-Castroviejo D, López LC. CoQ deficiency causes disruption of mitochondrial sulfide oxidation, a new pathomechanism associated with this syndrome. EMBO Mol Med 2017; 9:78-95. [PMID: 27856619 PMCID: PMC5210161 DOI: 10.15252/emmm.201606345] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Coenzyme Q (CoQ) is a key component of the mitochondrial respiratory chain, but it also has several other functions in the cellular metabolism. One of them is to function as an electron carrier in the reaction catalyzed by sulfide:quinone oxidoreductase (SQR), which catalyzes the first reaction in the hydrogen sulfide oxidation pathway. Therefore, SQR may be affected by CoQ deficiency. Using human skin fibroblasts and two mouse models with primary CoQ deficiency, we demonstrate that severe CoQ deficiency causes a reduction in SQR levels and activity, which leads to an alteration of mitochondrial sulfide metabolism. In cerebrum of Coq9R239X mice, the deficit in SQR induces an increase in thiosulfate sulfurtransferase and sulfite oxidase, as well as modifications in the levels of thiols. As a result, biosynthetic pathways of glutamate, serotonin, and catecholamines were altered in the cerebrum, and the blood pressure was reduced. Therefore, this study reveals the reduction in SQR activity as one of the pathomechanisms associated with CoQ deficiency syndrome.
Collapse
Affiliation(s)
- Marta Luna-Sánchez
- Departmento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain .,Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Agustín Hidalgo-Gutiérrez
- Departmento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain.,Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | | | - Julio Chaves-Serrano
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Eliana Barriocanal-Casado
- Departmento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain.,Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | | | - Miguel Romero
- Departmento de Farmacología, Facultad de Farmacia, Instituto de Investigación Biosanitaria de Granada, Universidad de Granada, Granada, Spain
| | - Ramy Ka Sayed
- Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Juan Duarte
- Departmento de Farmacología, Facultad de Farmacia, Instituto de Investigación Biosanitaria de Granada, Universidad de Granada, Granada, Spain
| | - Holger Prokisch
- Institute of Human Genetics, Technische Universität München, München, Germany
| | - Markus Schuelke
- Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Distelmaier
- Department of General Pediatrics, Heinrich-Heine-University, Düsseldorf, Germany
| | - Germaine Escames
- Departmento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain.,Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Darío Acuña-Castroviejo
- Departmento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain.,Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Luis C López
- Departmento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain .,Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| |
Collapse
|
41
|
Stefely JA, Pagliarini DJ. Biochemistry of Mitochondrial Coenzyme Q Biosynthesis. Trends Biochem Sci 2017; 42:824-843. [PMID: 28927698 DOI: 10.1016/j.tibs.2017.06.008] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/14/2017] [Accepted: 06/22/2017] [Indexed: 11/16/2022]
Abstract
Coenzyme Q (CoQ, ubiquinone) is a redox-active lipid produced across all domains of life that functions in electron transport and oxidative phosphorylation and whose deficiency causes human diseases. Yet, CoQ biosynthesis has not been fully defined in any organism. Several proteins with unclear molecular functions facilitate CoQ biosynthesis through unknown means, and multiple steps in the pathway are catalyzed by currently unidentified enzymes. Here we highlight recent progress toward filling these knowledge gaps through both traditional biochemistry and cutting-edge 'omics' approaches. To help fill the remaining gaps, we present questions framed by the recently discovered CoQ biosynthetic complex and by putative biophysical barriers. Mapping CoQ biosynthesis, metabolism, and transport pathways has great potential to enhance treatment of numerous human diseases.
Collapse
Affiliation(s)
- Jonathan A Stefely
- Morgridge Institute for Research, Madison, WI, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA; School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Pagliarini
- Morgridge Institute for Research, Madison, WI, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
42
|
Pierce JD, Shen Q, Peltzer J, Thimmesch A, Hiebert JB. A pilot study exploring the effects of ubiquinol on brain genomics after traumatic brain injury. Nurs Outlook 2017; 65:S44-S52. [DOI: 10.1016/j.outlook.2017.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 12/14/2022]
|
43
|
Pierrel F. Impact of Chemical Analogs of 4-Hydroxybenzoic Acid on Coenzyme Q Biosynthesis: From Inhibition to Bypass of Coenzyme Q Deficiency. Front Physiol 2017; 8:436. [PMID: 28690551 PMCID: PMC5479927 DOI: 10.3389/fphys.2017.00436] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/08/2017] [Indexed: 12/21/2022] Open
Abstract
Coenzyme Q is a lipid that participates to important physiological functions. Coenzyme Q is synthesized in multiple steps from the precursor 4-hydroxybenzoic acid. Mutations in enzymes that participate to coenzyme Q biosynthesis result in primary coenzyme Q deficiency, a type of mitochondrial disease. Coenzyme Q10 supplementation of patients is the classical treatment but it shows limited efficacy in some cases. The molecular understanding of the coenzyme Q biosynthetic pathway allowed the design of experiments to bypass deficient biosynthetic steps with analogs of 4-hydroxybenzoic acid. These molecules provide the defective chemical group and can reactivate endogenous coenzyme Q biosynthesis as demonstrated recently in yeast, mammalian cell cultures, and mouse models of primary coenzyme Q deficiency. This mini review presents how the chemical properties of various analogs of 4-hydroxybenzoic acid dictate the effect of the molecules on CoQ biosynthesis and how the reactivation of endogenous coenzyme Q biosynthesis may achieve better results than exogenous CoQ10 supplementation.
Collapse
Affiliation(s)
- Fabien Pierrel
- Centre National de la Recherche Scientifique, Grenoble INP, TIMC-IMAG, University Grenoble AlpesGrenoble, France
| |
Collapse
|
44
|
Neergheen V, Chalasani A, Wainwright L, Yubero D, Montero R, Artuch R, Hargreaves I. Coenzyme Q10 in the Treatment of Mitochondrial Disease. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2017. [DOI: 10.1177/2326409817707771] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Viruna Neergheen
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Annapurna Chalasani
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Luke Wainwright
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Delia Yubero
- Clinical Biochemistry department, Institut de Recerca Sant Joan de Déu and CIBERER, Barcelona, Spain
| | - Raquel Montero
- Clinical Biochemistry department, Institut de Recerca Sant Joan de Déu and CIBERER, Barcelona, Spain
| | - Rafael Artuch
- Clinical Biochemistry department, Institut de Recerca Sant Joan de Déu and CIBERER, Barcelona, Spain
| | - Iain Hargreaves
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
- School of Pharmacy and Biomolecular Science, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
45
|
|
46
|
Peng X, Xing P, Li X, Qian Y, Song F, Bai Z, Han G, Lei H. Towards Personalized Intervention for Alzheimer's Disease. GENOMICS PROTEOMICS & BIOINFORMATICS 2016; 14:289-297. [PMID: 27693548 PMCID: PMC5093853 DOI: 10.1016/j.gpb.2016.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/14/2016] [Accepted: 01/31/2016] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) remains to be a grand challenge for the international community despite over a century of exploration. A key factor likely accounting for such a situation is the vast heterogeneity in the disease etiology, which involves very complex and divergent pathways. Therefore, intervention strategies shall be tailored for subgroups of AD patients. Both demographic and in-depth information is needed for patient stratification. The demographic information includes primarily APOE genotype, age, gender, education, environmental exposure, life style, and medical history, whereas in-depth information stems from genome sequencing, brain imaging, peripheral biomarkers, and even functional assays on neurons derived from patient-specific induced pluripotent cells (iPSCs). Comprehensive information collection, better understanding of the disease mechanisms, and diversified strategies of drug development would help with more effective intervention in the foreseeable future.
Collapse
Affiliation(s)
- Xing Peng
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peiqi Xing
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiuhui Li
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Qian
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fuhai Song
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhouxian Bai
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangchun Han
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongxing Lei
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Cunji Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing 100053, China.
| |
Collapse
|
47
|
Acosta MJ, Vazquez Fonseca L, Desbats MA, Cerqua C, Zordan R, Trevisson E, Salviati L. Coenzyme Q biosynthesis in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:1079-1085. [PMID: 27060254 DOI: 10.1016/j.bbabio.2016.03.036] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 01/11/2023]
Abstract
Coenzyme Q (CoQ, or ubiquinone) is a remarkable lipid that plays an essential role in mitochondria as an electron shuttle between complexes I and II of the respiratory chain, and complex III. It is also a cofactor of other dehydrogenases, a modulator of the permeability transition pore and an essential antioxidant. CoQ is synthesized in mitochondria by a set of at least 12 proteins that form a multiprotein complex. The exact composition of this complex is still unclear. Most of the genes involved in CoQ biosynthesis (COQ genes) have been studied in yeast and have mammalian orthologues. Some of them encode enzymes involved in the modification of the quinone ring of CoQ, but for others the precise function is unknown. Two genes appear to have a regulatory role: COQ8 (and its human counterparts ADCK3 and ADCK4) encodes a putative kinase, while PTC7 encodes a phosphatase required for the activation of Coq7. Mutations in human COQ genes cause primary CoQ(10) deficiency, a clinically heterogeneous mitochondrial disorder with onset from birth to the seventh decade, and with clinical manifestation ranging from fatal multisystem disorders, to isolated encephalopathy or nephropathy. The pathogenesis of CoQ(10) deficiency involves deficient ATP production and excessive ROS formation, but possibly other aspects of CoQ(10) function are implicated. CoQ(10) deficiency is unique among mitochondrial disorders since an effective treatment is available. Many patients respond to oral CoQ(10) supplementation. Nevertheless, treatment is still problematic because of the low bioavailability of the compound, and novel pharmacological approaches are currently being investigated. This article is part of a Special Issue entitled 'EBEC 2016: 19th European Bioenergetics Conference, Riva del Garda, Italy, July 2-6, 2016', edited by Prof. Paolo Bernardi.
Collapse
Affiliation(s)
- Manuel Jesús Acosta
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, and IRP Città della Speranza, Padova, Italy
| | - Luis Vazquez Fonseca
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, and IRP Città della Speranza, Padova, Italy
| | - Maria Andrea Desbats
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, and IRP Città della Speranza, Padova, Italy
| | - Cristina Cerqua
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, and IRP Città della Speranza, Padova, Italy
| | - Roberta Zordan
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, and IRP Città della Speranza, Padova, Italy
| | - Eva Trevisson
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, and IRP Città della Speranza, Padova, Italy.
| | - Leonardo Salviati
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, and IRP Città della Speranza, Padova, Italy.
| |
Collapse
|
48
|
Luna-Sánchez M, Díaz-Casado E, Barca E, Tejada MÁ, Montilla-García Á, Cobos EJ, Escames G, Acuña-Castroviejo D, Quinzii CM, López LC. The clinical heterogeneity of coenzyme Q10 deficiency results from genotypic differences in the Coq9 gene. EMBO Mol Med 2016; 7:670-87. [PMID: 25802402 PMCID: PMC4492823 DOI: 10.15252/emmm.201404632] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Primary coenzyme Q10 (CoQ10) deficiency is due to mutations in genes involved in CoQ biosynthesis. The disease has been associated with five major phenotypes, but a genotype-phenotype correlation is unclear. Here, we compare two mouse models with a genetic modification in Coq9 gene (Coq9(Q95X) and Coq9(R239X)), and their responses to 2,4-dihydroxybenzoic acid (2,4-diHB). Coq9(R239X) mice manifest severe widespread CoQ deficiency associated with fatal encephalomyopathy and respond to 2,4-diHB increasing CoQ levels. In contrast, Coq9(Q95X) mice exhibit mild CoQ deficiency manifesting with reduction in CI+III activity and mitochondrial respiration in skeletal muscle, and late-onset mild mitochondrial myopathy, which does not respond to 2,4-diHB. We show that these differences are due to the levels of COQ biosynthetic proteins, suggesting that the presence of a truncated version of COQ9 protein in Coq9(R239X) mice destabilizes the CoQ multiprotein complex. Our study points out the importance of the multiprotein complex for CoQ biosynthesis in mammals, which may provide new insights to understand the genotype-phenotype heterogeneity associated with human CoQ deficiency and may have a potential impact on the treatment of this mitochondrial disorder.
Collapse
Affiliation(s)
- Marta Luna-Sánchez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain Centro de Investigación Biomédica, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| | - Elena Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain Centro de Investigación Biomédica, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| | - Emanuele Barca
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Miguel Ángel Tejada
- Departamento de Farmacología, Facultad de Medicina, Universidad de Granada, Granada, Spain Centro de Investigación Biomédica, Instituto de Neurociencias, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| | - Ángeles Montilla-García
- Departamento de Farmacología, Facultad de Medicina, Universidad de Granada, Granada, Spain Centro de Investigación Biomédica, Instituto de Neurociencias, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| | - Enrique Javier Cobos
- Departamento de Farmacología, Facultad de Medicina, Universidad de Granada, Granada, Spain Centro de Investigación Biomédica, Instituto de Neurociencias, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| | - Germaine Escames
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain Centro de Investigación Biomédica, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| | - Dario Acuña-Castroviejo
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain Centro de Investigación Biomédica, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| | - Catarina M Quinzii
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Luis Carlos López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain Centro de Investigación Biomédica, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| |
Collapse
|
49
|
Fragaki K, Chaussenot A, Benoist JF, Ait-El-Mkadem S, Bannwarth S, Rouzier C, Cochaud C, Paquis-Flucklinger V. Coenzyme Q10 defects may be associated with a deficiency of Q10-independent mitochondrial respiratory chain complexes. Biol Res 2016; 49:4. [PMID: 26742794 PMCID: PMC4705639 DOI: 10.1186/s40659-015-0065-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/30/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Coenzyme Q10 (CoQ10 or ubiquinone) deficiency can be due either to mutations in genes involved in CoQ10 biosynthesis pathway, or to mutations in genes unrelated to CoQ10 biosynthesis. CoQ10 defect is the only oxidative phosphorylation disorder that can be clinically improved after oral CoQ10 supplementation. Thus, early diagnosis, first evoked by mitochondrial respiratory chain (MRC) spectrophotometric analysis, then confirmed by direct measurement of CoQ10 levels, is of critical importance to prevent irreversible damage in organs such as the kidney and the central nervous system. It is widely reported that CoQ10 deficient patients present decreased quinone-dependent activities (segments I + III or G3P + III and II + III) while MRC activities of complexes I, II, III, IV and V are normal. We previously suggested that CoQ10 defect may be associated with a deficiency of CoQ10-independent MRC complexes. The aim of this study was to verify this hypothesis in order to improve the diagnosis of this disease. RESULTS To determine whether CoQ10 defect could be associated with MRC deficiency, we quantified CoQ10 by LC-MSMS in a cohort of 18 patients presenting CoQ10-dependent deficiency associated with MRC defect. We found decreased levels of CoQ10 in eight patients out of 18 (45 %), thus confirming CoQ10 disease. CONCLUSIONS Our study shows that CoQ10 defect can be associated with MRC deficiency. This could be of major importance in clinical practice for the diagnosis of a disease that can be improved by CoQ10 supplementation.
Collapse
Affiliation(s)
- Konstantina Fragaki
- School of Medicine, IRCAN, UMR CNRS 7284/INSERM U1081/UNS, Nice Sophia-Antipolis University, 28 av de Valombrose, 06107, Nice Cedex 2, France. .,Department of Medical Genetics, Nice Teaching Hospital, National Centre for Mitochondrial Diseases, Nice, France.
| | - Annabelle Chaussenot
- School of Medicine, IRCAN, UMR CNRS 7284/INSERM U1081/UNS, Nice Sophia-Antipolis University, 28 av de Valombrose, 06107, Nice Cedex 2, France. .,Department of Medical Genetics, Nice Teaching Hospital, National Centre for Mitochondrial Diseases, Nice, France.
| | | | - Samira Ait-El-Mkadem
- School of Medicine, IRCAN, UMR CNRS 7284/INSERM U1081/UNS, Nice Sophia-Antipolis University, 28 av de Valombrose, 06107, Nice Cedex 2, France. .,Department of Medical Genetics, Nice Teaching Hospital, National Centre for Mitochondrial Diseases, Nice, France.
| | - Sylvie Bannwarth
- School of Medicine, IRCAN, UMR CNRS 7284/INSERM U1081/UNS, Nice Sophia-Antipolis University, 28 av de Valombrose, 06107, Nice Cedex 2, France. .,Department of Medical Genetics, Nice Teaching Hospital, National Centre for Mitochondrial Diseases, Nice, France.
| | - Cécile Rouzier
- School of Medicine, IRCAN, UMR CNRS 7284/INSERM U1081/UNS, Nice Sophia-Antipolis University, 28 av de Valombrose, 06107, Nice Cedex 2, France. .,Department of Medical Genetics, Nice Teaching Hospital, National Centre for Mitochondrial Diseases, Nice, France.
| | - Charlotte Cochaud
- School of Medicine, IRCAN, UMR CNRS 7284/INSERM U1081/UNS, Nice Sophia-Antipolis University, 28 av de Valombrose, 06107, Nice Cedex 2, France.
| | - Véronique Paquis-Flucklinger
- School of Medicine, IRCAN, UMR CNRS 7284/INSERM U1081/UNS, Nice Sophia-Antipolis University, 28 av de Valombrose, 06107, Nice Cedex 2, France. .,Department of Medical Genetics, Nice Teaching Hospital, National Centre for Mitochondrial Diseases, Nice, France.
| |
Collapse
|
50
|
Chung WK, Martin K, Jalas C, Braddock SR, Juusola J, Monaghan KG, Warner B, Franks S, Yudkoff M, Lulis L, Rhodes RH, Prasad V, Torti E, Cho MT, Shinawi M. Mutations inCOQ4, an essential component of coenzyme Q biosynthesis, cause lethal neonatal mitochondrial encephalomyopathy. J Med Genet 2015; 52:627-35. [DOI: 10.1136/jmedgenet-2015-103140] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/17/2015] [Indexed: 12/16/2022]
|