1
|
Hernández‐Camacho JD, Vicente‐García C, Ardila‐García L, Padilla‐Campos A, López‐Lluch G, Santos‐Ocaña C, Zammit PS, Carvajal JJ, Navas P, Fernández‐Ayala DJ. Prenatal and progressive coenzyme Q 10 administration to mitigate muscle dysfunction in mitochondrial disease. J Cachexia Sarcopenia Muscle 2024; 15:2402-2416. [PMID: 39354863 PMCID: PMC11634497 DOI: 10.1002/jcsm.13574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND ADCK genes encode aarF domain-containing mitochondrial kinases involved in coenzyme Q (CoQ) biosynthesis and regulation. Haploinsufficiency of ADCK2 in humans leads to adult-onset physical incapacity with reduced mitochondrial CoQ levels in skeletal muscle, resulting in mitochondrial myopathy and alterations in fatty acid β-oxidation. The sole current treatment for CoQ deficiencies is oral administration of CoQ10, which causes only partial recovery with postnatal treatment, underscoring the importance of early diagnosis for successful intervention. METHODS We used Adck2 heterozygous mice to examine the influence of this gene on muscle structure, function and regeneration throughout development, growth and ageing. This investigation involved techniques including immunohistochemistry, analysis of CoQ levels, mitochondrial respiratory content, muscle transcriptome analysis and functional tests. RESULTS We demonstrated that Adck2 heterozygous mice exhibit defects from embryonic development, particularly in skeletal muscle (1102 genes deregulated). Adck2 heterozygous embryos were 7% smaller in size and displayed signs of delayed development. Prenatal administration of CoQ10 could mitigate these embryonic defects. Heterozygous Adck2 mice also showed a decrease in myogenic cell differentiation, with more severe consequences in 'aged' mice (41.63% smaller) (P < 0.01). Consequently, heterozygous Adck2 mice displayed accelerated muscle wasting associated with ageing in muscle structure (P < 0.05), muscle function (less grip strength capacity) (P < 0.001) and muscle mitochondrial respiration (P < 0.001). Furthermore, progressive CoQ10 administration conferred protective effects on mitochondrial function (P < 0.0001) and skeletal muscle (P < 0.05). CONCLUSIONS Our work uncovered novel aspects of CoQ deficiencies, revealing defects during embryonic development in mammals for the first time. Additionally, we identified the gradual establishment and progression of the deleterious Adck2 mouse phenotype. Importantly, CoQ10 supplementation demonstrated a protective effect when initiated during development.
Collapse
Affiliation(s)
- Juan Diego Hernández‐Camacho
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
- CIBERERInstituto de Salud Carlos IIIMadridSpain
| | | | - Lorena Ardila‐García
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
| | - Ana Padilla‐Campos
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
| | - Guillermo López‐Lluch
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
- CIBERERInstituto de Salud Carlos IIIMadridSpain
| | - Carlos Santos‐Ocaña
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
- CIBERERInstituto de Salud Carlos IIIMadridSpain
| | - Peter S. Zammit
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - Jaime J. Carvajal
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
| | - Plácido Navas
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
- CIBERERInstituto de Salud Carlos IIIMadridSpain
| | - Daniel J.M. Fernández‐Ayala
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
- CIBERERInstituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
2
|
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
|
3
|
Díaz-Casado ME, González-García P, López-Herrador S, Hidalgo-Gutiérrez A, Jiménez-Sánchez L, Barriocanal-Casado E, Bakkali M, van de Lest CHA, Corral-Sarasa J, Zaal EA, Berkers CR, López LC. Oral β-RA induces metabolic rewiring leading to the rescue of diet-induced obesity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167283. [PMID: 38851305 DOI: 10.1016/j.bbadis.2024.167283] [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: 03/25/2024] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Obesity represents a significant health challenge, intricately linked to conditions such as type II diabetes, metabolic syndrome, and hepatic steatosis. Several existing obesity treatments exhibit limited efficacy, undesirable side effects or a limited capability to maintain therapeutics effects in the long-term. Recently, modulation Coenzyme Q (CoQ) metabolism has emerged as a promising target for treatment of metabolic syndrome. This potential intervention could involve the modulation of endogenous CoQ biosynthesis by the use of analogs of the precursor of its biosynthesis, such as β-resorcylic acid (β-RA). Here, we show that oral supplementation with β-RA, incorporated into the diet of diet-induced obese (DIO) mice, leads to substantial weight loss. The anti-obesity effects of β-RA are partially elucidated through the normalization of mitochondrial CoQ metabolism in white adipose tissue (WAT). Additionally, we identify an HFN4α/LXR-dependent transcriptomic activation of the hepatic lipid metabolism that contributes to the anti-obesity effects of β-RA. Consequently, β-RA mitigates WAT hypertrophy, prevents hepatic steatosis, counteracts metabolic abnormalities in WAT and liver, and enhances glucose homeostasis by reducing the insulin/glucagon ratio and plasma levels of gastric inhibitory peptide (GIP). Moreover, pharmacokinetic evaluation of β-RA supports its translational potential. Thus, β-RA emerges as an efficient, safe, and translatable therapeutic option for the treatment and/or prevention of obesity, metabolic dysfunction-associated steatotic liver disease (MASLD).
Collapse
Affiliation(s)
- 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; Instituto de Investigación Biosanitaria Ibs. Granada, 18016 Granada, Spain
| | - 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; Instituto de Investigación Biosanitaria Ibs. Granada, 18016 Granada, Spain
| | - Sergio López-Herrador
- 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; Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | | | - Eliana Barriocanal-Casado
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA; GENYO, Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer-University of Granada-Andalusian Regional Government, 18016 Granada, Spain
| | - Mohammed Bakkali
- Departamento de Genética, Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain
| | - Chris H A van de Lest
- Division of Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3508 TD Utrecht, the Netherlands
| | | | - Esther A Zaal
- 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
- Division of Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3508 TD 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; Instituto de Investigación Biosanitaria Ibs. Granada, 18016 Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), 18016 Granada, Spain.
| |
Collapse
|
4
|
Fabra MA, Paredes-Fuentes AJ, Torralba Carnerero M, Moreno Férnandez de Ayala DJ, Arroyo Luque A, Sánchez Cuesta A, Staiano C, Sanchez-Pintos P, Luz Couce M, Tomás M, Marco-Hernández AV, Orellana C, Martínez F, Roselló M, Caro A, Oltra Soler JS, Monfort S, Sánchez A, Rausell D, Vitoria I, Del Toro M, Garcia-Cazorla A, Julia-Palacios NA, Jou C, Yubero D, López LC, Hernández Camacho JD, López Lluch G, Ballesteros Simarro M, Rodríguez Aguilera JC, Calvo GB, Cascajo Almenara MV, Artuch R, Santos-Ocaña C. New variants expand the neurological phenotype of COQ7 deficiency. J Inherit Metab Dis 2024; 47:1047-1068. [PMID: 38973597 DOI: 10.1002/jimd.12776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 07/09/2024]
Abstract
The protein encoded by COQ7 is required for CoQ10 synthesis in humans, hydroxylating 3-demethoxyubiquinol (DMQ10) in the second to last steps of the pathway. COQ7 mutations lead to a primary CoQ10 deficiency syndrome associated with a pleiotropic neurological disorder. This study shows the clinical, physiological, and molecular characterization of four new cases of CoQ10 primary deficiency caused by five mutations in COQ7, three of which have not yet been described, inducing mitochondrial dysfunction in all patients. However, the specific combination of the identified variants in each patient generated precise pathophysiological and molecular alterations in fibroblasts, which would explain the differential in vitro response to supplementation therapy. Our results suggest that COQ7 dysfunction could be caused by specific structural changes that affect the interaction with COQ9 required for the DMQ10 presentation to COQ7, the substrate access to the active site, and the maintenance of the active site structure. Remarkably, patients' fibroblasts share transcriptional remodeling, supporting a modification of energy metabolism towards glycolysis, which could be an adaptive mechanism against CoQ10 deficiency. However, transcriptional analysis of mitochondria-associated pathways showed distinct and dramatic differences between patient fibroblasts, which correlated with the extent of pathophysiological and neurological alterations observed in the probands. Overall, this study suggests that the combination of precise genetic diagnostics and the availability of new structural models of human proteins could help explain the origin of phenotypic pleiotropy observed in some genetic diseases and the different responses to available therapies.
Collapse
Affiliation(s)
- María Alcázar Fabra
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Abraham J Paredes-Fuentes
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Manuel Torralba Carnerero
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
| | - Daniel J Moreno Férnandez de Ayala
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Antonio Arroyo Luque
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Ana Sánchez Cuesta
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Carmine Staiano
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Paula Sanchez-Pintos
- Unidad de Diagnóstico y Tratamiento de Enfermedades Metabólicas Congénitas, Hospital de Santiago de Compostela, IDIS, CIBERER, MetabERN, Santiago de Compostela, Spain
- GCV14/ER/5 CIBERER, Instituto de Salud Carlos III, Santiago de Compostela, Spain
| | - María Luz Couce
- Unidad de Diagnóstico y Tratamiento de Enfermedades Metabólicas Congénitas, Hospital de Santiago de Compostela, IDIS, CIBERER, MetabERN, Santiago de Compostela, Spain
- GCV14/ER/5 CIBERER, Instituto de Salud Carlos III, Santiago de Compostela, Spain
| | - Miguel Tomás
- Hospital Universitari i Politècnic La Fe, Servicio de Neuropediatría, Valencia, Spain
| | | | - Carmen Orellana
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | - Francisco Martínez
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | - Mónica Roselló
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | - Alfonso Caro
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | | | - Sandra Monfort
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | - Alejandro Sánchez
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | - Dolores Rausell
- Hospital Universitari i Politècnic La Fe, Servicio de Análisis Clínicos, Valencia, Spain
| | - Isidro Vitoria
- Hospital Universitari i Politècnic La Fe, Unidad de Metabolopatías, Valencia, Spain
| | - Mireia Del Toro
- Pediatric Neurology Unit, Hospital Universitari Vall d'Hebron, CIBERER, MetabERN, Barcelona, Spain
- Instituto de Salud Carlos III, Barcelona, Spain
| | - Angels Garcia-Cazorla
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Natalia A Julia-Palacios
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Cristina Jou
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Delia Yubero
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Luis Carlos López
- Departamento de Fisiología, Facultad de Medicina, Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, Granada, Spain
| | - Juan Diego Hernández Camacho
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Guillermo López Lluch
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Manuel Ballesteros Simarro
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Juan Carlos Rodríguez Aguilera
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Gloria Brea Calvo
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - María Victoria Cascajo Almenara
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Rafael Artuch
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Carlos Santos-Ocaña
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| |
Collapse
|
5
|
Yu W, Wu W, Zhao D, Zhang R, Shao K, Liu H, Yan C, Lin P. Idebenone ameliorates statin-induced myotoxicity in atherosclerotic ApoE-/- mice by reducing oxidative stress and improving mitochondrial function. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167157. [PMID: 38582266 DOI: 10.1016/j.bbadis.2024.167157] [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: 10/20/2023] [Revised: 03/23/2024] [Accepted: 03/30/2024] [Indexed: 04/08/2024]
Abstract
Statins are the first line of choice for the treatment for atherosclerosis, but their use can cause myotoxicity, a common side effect that may require dosage reduction or discontinuation. The exact mechanism of statin-induced myotoxicity is unknown. Previous research has demonstrated that the combination of idebenone and statin yielded superior anti-atherosclerotic outcomes. Here, we investigated the mechanism of statin-induced myotoxicity in atherosclerotic ApoE-/- mice and whether idebenone could counteract it. After administering simvastatin to ApoE-/- mice, we observed a reduction in plaque formation as well as a decrease in their exercise capacity. We observed elevated levels of lactic acid and creatine kinase, along with a reduction in the cross-sectional area of muscle fibers, an increased presence of ragged red fibers, heightened mitochondrial crista lysis, impaired mitochondrial complex activity, and decreased levels of CoQ9 and CoQ10. Two-photon fluorescence imaging revealed elevated H2O2 levels in the quadriceps, indicating increased oxidative stress. Proteomic analysis indicated that simvastatin inhibited the tricarboxylic acid cycle. Idebenone treatment not only further reduced plaque formation but also ameliorated the impaired exercise capacity caused by simvastatin. Our study represents the inaugural comprehensive investigation into the mechanisms underlying statin-induced myotoxicity. We have demonstrated that statins inhibit CoQ synthesis, impair mitochondrial complex functionality, and elevate oxidative stress, ultimately resulting in myotoxic effects. Furthermore, our research marks the pioneering identification of idebenone's capability to mitigate statin-induced myotoxicity by attenuating oxidative stress, thereby safeguarding mitochondrial complex functionality. The synergistic use of idebenone and statin not only enhances the effectiveness against atherosclerosis but also mitigates statin-induced myotoxicity.
Collapse
Affiliation(s)
- Wenfei Yu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China; University of Health and Rehabilitation Sciences, No. 17, Shandong Road, Shinan district, Qingdao City, Shandong Province, China
| | - Wenjing Wu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Dandan Zhao
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Rui Zhang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Kai Shao
- Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong 266000, China
| | - Haoyang Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Chuanzhu Yan
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Pengfei Lin
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China.
| |
Collapse
|
6
|
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
|
7
|
Guile MD, Jain A, Anderson KA, Clarke CF. New Insights on the Uptake and Trafficking of Coenzyme Q. Antioxidants (Basel) 2023; 12:1391. [PMID: 37507930 PMCID: PMC10376127 DOI: 10.3390/antiox12071391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/30/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Coenzyme Q (CoQ) is an essential lipid with many cellular functions, such as electron transport for cellular respiration, antioxidant protection, redox homeostasis, and ferroptosis suppression. Deficiencies in CoQ due to aging, genetic disease, or medication can be ameliorated by high-dose supplementation. As such, an understanding of the uptake and transport of CoQ may inform methods of clinical use and identify how to better treat deficiency. Here, we review what is known about the cellular uptake and intracellular distribution of CoQ from yeast, mammalian cell culture, and rodent models, as well as its absorption at the organism level. We discuss the use of these model organisms to probe the mechanisms of uptake and distribution. The literature indicates that CoQ uptake and distribution are multifaceted processes likely to have redundancies in its transport, utilizing the endomembrane system and newly identified proteins that function as lipid transporters. Impairment of the trafficking of either endogenous or exogenous CoQ exerts profound effects on metabolism and stress response. This review also highlights significant gaps in our knowledge of how CoQ is distributed within the cell and suggests future directions of research to better understand this process.
Collapse
Affiliation(s)
- Michael D Guile
- Department of Chemistry & Biochemistry and the Molecular Biology Institute, University of California, Los Angeles, CA 90059, USA
| | - Akash Jain
- Department of Chemistry & Biochemistry and the Molecular Biology Institute, University of California, Los Angeles, CA 90059, USA
| | - Kyle A Anderson
- Department of Chemistry & Biochemistry and the Molecular Biology Institute, University of California, Los Angeles, CA 90059, USA
| | - Catherine F Clarke
- Department of Chemistry & Biochemistry and the Molecular Biology Institute, University of California, Los Angeles, CA 90059, USA
| |
Collapse
|
8
|
Hu M, Jiang Y, Xu JJ. Characterization of Arabidopsis thaliana Coq9 in the CoQ Biosynthetic Pathway. Metabolites 2023; 13:813. [PMID: 37512520 PMCID: PMC10385794 DOI: 10.3390/metabo13070813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Coenzyme Q, also known as ubiquinone, is a fat-soluble isoprene quinone that serves as a cofactor for numerous enzymes across all domains of life. However, the biosynthetic pathway for this important molecule in plants has been examined in only a limited number of studies. In yeast and mammals, Coq9, an isoprenoid-lipid-binding protein, is essential for CoQ biosynthesis. Previous studies showed that Arabidopsis thaliana Coq9 failed to complement the fission yeast Schizosaccharomyces pombe coq9 null mutant, and its function in plants remains unknown. In this study, we demonstrated that expression of Arabidopsis Coq9 rescued the growth of a yeast temperature-sensitive coq9 mutant and increased CoQ content. Phylogenetic analysis revealed that Coq9 is widely present in green plants. Green fluorescent protein (GFP) fusion experiments showed that Arabidopsis Coq9 is targeted to mitochondria. Disruption of the Coq9 gene in Arabidopsis results in lower amounts of CoQ. Our work suggests that plant Coq9 is required for efficient CoQ biosynthesis. These findings provide new insights into the evolution of CoQ biosynthesis in plants. The identification of Coq9 as a key player in CoQ biosynthesis in plants opens up new avenues for understanding the regulation of this important metabolic pathway.
Collapse
Affiliation(s)
- Mei Hu
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Biology and the Environment, Nanjing Forestry University, Nanjing 210037, China
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai 201602, China
| | - Yan Jiang
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai 201602, China
- School of Life Sciences, Shanghai Normal University, Shanghai 200234, China
| | - Jing-Jing Xu
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai 201602, China
| |
Collapse
|
9
|
Olkhova EA, Smith LA, Bradshaw C, Gorman GS, Erskine D, Ng YS. Neurological Phenotypes in Mouse Models of Mitochondrial Disease and Relevance to Human Neuropathology. Int J Mol Sci 2023; 24:ijms24119698. [PMID: 37298649 DOI: 10.3390/ijms24119698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Mitochondrial diseases represent the most common inherited neurometabolic disorders, for which no effective therapy currently exists for most patients. The unmet clinical need requires a more comprehensive understanding of the disease mechanisms and the development of reliable and robust in vivo models that accurately recapitulate human disease. This review aims to summarise and discuss various mouse models harbouring transgenic impairments in genes that regulate mitochondrial function, specifically their neurological phenotype and neuropathological features. Ataxia secondary to cerebellar impairment is one of the most prevalent neurological features of mouse models of mitochondrial dysfunction, consistent with the observation that progressive cerebellar ataxia is a common neurological manifestation in patients with mitochondrial disease. The loss of Purkinje neurons is a shared neuropathological finding in human post-mortem tissues and numerous mouse models. However, none of the existing mouse models recapitulate other devastating neurological phenotypes, such as refractory focal seizures and stroke-like episodes seen in patients. Additionally, we discuss the roles of reactive astrogliosis and microglial reactivity, which may be driving the neuropathology in some of the mouse models of mitochondrial dysfunction, as well as mechanisms through which cellular death may occur, beyond apoptosis, in neurons undergoing mitochondrial bioenergy crisis.
Collapse
Affiliation(s)
- Elizaveta A Olkhova
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Laura A Smith
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Carla Bradshaw
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Gráinne S Gorman
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Daniel Erskine
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Yi Shiau Ng
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|
10
|
Manicki M, Aydin H, Abriata LA, Overmyer KA, Guerra RM, Coon JJ, Dal Peraro M, Frost A, Pagliarini DJ. Structure and functionality of a multimeric human COQ7:COQ9 complex. Mol Cell 2022; 82:4307-4323.e10. [PMID: 36306796 PMCID: PMC10058641 DOI: 10.1016/j.molcel.2022.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 07/01/2022] [Accepted: 10/04/2022] [Indexed: 11/18/2022]
Abstract
Coenzyme Q (CoQ) is a redox-active lipid essential for core metabolic pathways and antioxidant defense. CoQ is synthesized upon the mitochondrial inner membrane by an ill-defined "complex Q" metabolon. Here, we present structure-function analyses of a lipid-, substrate-, and NADH-bound complex comprising two complex Q subunits: the hydroxylase COQ7 and the lipid-binding protein COQ9. We reveal that COQ7 adopts a ferritin-like fold with a hydrophobic channel whose substrate-binding capacity is enhanced by COQ9. Using molecular dynamics, we further show that two COQ7:COQ9 heterodimers form a curved tetramer that deforms the membrane, potentially opening a pathway for the CoQ intermediates to translocate from the bilayer to the proteins' lipid-binding sites. Two such tetramers assemble into a soluble octamer with a pseudo-bilayer of lipids captured within. Together, these observations indicate that COQ7 and COQ9 cooperate to access hydrophobic precursors within the membrane and coordinate subsequent synthesis steps toward producing CoQ.
Collapse
Affiliation(s)
- Mateusz Manicki
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Morgridge Institute for Research, Madison, WI 53715, USA
| | - Halil Aydin
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Luciano A Abriata
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Protein Production and Structure Core Facility, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Katherine A Overmyer
- Morgridge Institute for Research, Madison, WI 53715, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53562, USA
| | - Rachel M Guerra
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Morgridge Institute for Research, Madison, WI 53715, USA
| | - Joshua J Coon
- Morgridge Institute for Research, Madison, WI 53715, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53506, USA
| | - Matteo Dal Peraro
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Adam Frost
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub and Altos Labs Bay Area Institute of Science, San Francisco, CA, USA.
| | - David J Pagliarini
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Morgridge Institute for Research, Madison, WI 53715, USA; Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
11
|
Banerjee R, Purhonen J, Kallijärvi J. The mitochondrial coenzyme Q junction and complex III: biochemistry and pathophysiology. FEBS J 2022; 289:6936-6958. [PMID: 34428349 DOI: 10.1111/febs.16164] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/13/2021] [Accepted: 08/23/2021] [Indexed: 01/13/2023]
Abstract
Coenzyme Q (CoQ, ubiquinone) is the electron-carrying lipid in the mitochondrial electron transport system (ETS). In mammals, it serves as the electron acceptor for nine mitochondrial inner membrane dehydrogenases. These include the NADH dehydrogenase (complex I, CI) and succinate dehydrogenase (complex II, CII) but also several others that are often omitted in the context of respiratory enzymes: dihydroorotate dehydrogenase, choline dehydrogenase, electron-transferring flavoprotein dehydrogenase, mitochondrial glycerol-3-phosphate dehydrogenase, proline dehydrogenases 1 and 2, and sulfide:quinone oxidoreductase. The metabolic pathways these enzymes are involved in range from amino acid and fatty acid oxidation to nucleotide biosynthesis, methylation, and hydrogen sulfide detoxification, among many others. The CoQ-linked metabolism depends on CoQ reoxidation by the mitochondrial complex III (cytochrome bc1 complex, CIII). However, the literature is surprisingly limited as for the role of the CoQ-linked metabolism in the pathogenesis of human diseases of oxidative phosphorylation (OXPHOS), in which the CoQ homeostasis is directly or indirectly affected. In this review, we give an introduction to CIII function, and an overview of the pathological consequences of CIII dysfunction in humans and mice and of the CoQ-dependent metabolic processes potentially affected in these pathological states. Finally, we discuss some experimental tools to dissect the various aspects of compromised CoQ oxidation.
Collapse
Affiliation(s)
- Rishi Banerjee
- Folkhälsan Research Center, Helsinki, Finland.,Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Janne Purhonen
- Folkhälsan Research Center, Helsinki, Finland.,Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Jukka Kallijärvi
- Folkhälsan Research Center, Helsinki, Finland.,Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Finland
| |
Collapse
|
12
|
Wang Y, Hekimi S. The CoQ biosynthetic di-iron carboxylate hydroxylase COQ7 is inhibited by in vivo metalation with manganese but remains functional by metalation with cobalt. MICROPUBLICATION BIOLOGY 2022; 2022:10.17912/micropub.biology.000635. [PMID: 36176269 PMCID: PMC9513594 DOI: 10.17912/micropub.biology.000635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/30/2022] [Accepted: 09/12/2022] [Indexed: 11/28/2022]
Abstract
Coenzyme Q (CoQ; ubiquinone) is an obligate component of the mitochondrial electron transport chain. COQ7 is a mitochondrial hydroxylase that is required for CoQ biosynthesis. COQ7 belongs to di-iron carboxylate enzymes, a rare type of enzyme that carries out a wide range of reactions. We found that manganese exposure of mouse cells leads to decreased COQ7 activity, but that pre-treatment with cobalt interferes with the inhibition by manganese. Our findings suggest that cobalt has greater affinity for the active site of COQ7 than both iron and manganese and that replacement of iron by cobalt at the active site preserves catalytic activity.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | - Siegfried Hekimi
- Department of Biology, McGill University, Montreal, Quebec, Canada
,
Correspondence to: Siegfried Hekimi (
)
| |
Collapse
|
13
|
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: 21] [Impact Index Per Article: 7.0] [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
|
14
|
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
|
15
|
Murray NH, Lewis A, Rincon Pabon JP, Gross ML, Henzler-Wildman K, Pagliarini DJ. 2-Propylphenol Allosterically Modulates COQ8A to Enhance ATPase Activity. ACS Chem Biol 2022; 17:2031-2038. [PMID: 35904798 PMCID: PMC9586199 DOI: 10.1021/acschembio.2c00434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
COQ8A is an atypical kinase-like protein that aids the biosynthesis of coenzyme Q, an essential cellular cofactor and antioxidant. COQ8A's mode of action remains unclear, in part due to the lack of small molecule tools to probe its function. Here, we blend NMR and hydrogen-deuterium exchange mass spectrometry to help determine how a small CoQ precursor mimetic, 2-propylphenol, modulates COQ8A activity. We identify a likely 2-propylphenol binding site and reveal that this compound modulates a conserved COQ8A domain to increase nucleotide affinity and ATPase activity. Our findings promise to aid further investigations into COQ8A's precise enzymatic function and the design of compounds capable of boosting endogenous CoQ production for therapeutic gain.
Collapse
Affiliation(s)
- Nathan H. Murray
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Adam Lewis
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Juan P. Rincon Pabon
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | - Michael L. Gross
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | | | - David J. Pagliarini
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Correspondence and requests for materials should be addressed to D.J.P.,
| |
Collapse
|
16
|
A novel COQ7 mutation causing primarily neuromuscular pathology and its treatment options. Mol Genet Metab Rep 2022; 31:100877. [PMID: 35782625 PMCID: PMC9248208 DOI: 10.1016/j.ymgmr.2022.100877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 11/21/2022] Open
Abstract
Coenzyme Q10 (CoQ10) is necessary as electron transporter in mitochondrial respiration and other cellular functions. CoQ10 is synthesized by all cells and defects in the synthesis pathway result in primary CoQ10 deficiency that frequently leads to severe mitochondrial disease syndrome. CoQ10 is exceedingly hydrophobic, insoluble, and poorly bioavailable, with the result that dietary CoQ10 supplementation produces no or only minimal relief for patients. We studied a patient from Turkey and identified and characterized a new mutation in the CoQ10 biosynthetic gene COQ7 (c.161G > A; p.Arg54Gln). We find that unexpected neuromuscular pathology can accompany CoQ10 deficiency caused by a COQ7 mutation. We also show that by-passing the need for COQ7 by providing the unnatural precursor 2,4-dihydroxybenzoic acid, as has been proposed, is unlikely to be an effective and safe therapeutic option. In contrast, we show for the first time in human patient cells that the respiratory defect resulting from CoQ10 deficiency is rescued by providing CoQ10 formulated with caspofungin (CF/CoQ). Caspofungin is a clinically approved intravenous fungicide whose surfactant properties lead to CoQ10 micellization, complete water solubilization, and efficient uptake by cells and organs in animal studies. These findings reinforce the possibility of using CF/CoQ in the clinical treatment of CoQ10-deficient patients.
Collapse
|
17
|
Primary coenzyme Q10 nephropathy, a potentially treatable form of steroid-resistant nephrotic syndrome. Pediatr Nephrol 2021; 36:3515-3527. [PMID: 33479824 PMCID: PMC8295399 DOI: 10.1007/s00467-020-04914-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/28/2020] [Accepted: 12/31/2020] [Indexed: 12/18/2022]
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is a genetically heterogeneous kidney disease that is the second most frequent cause of kidney failure in the first 2 decades of life. Despite the identification of mutations in more than 39 genes as causing SRNS, and the localization of its pathogenesis to glomerular podocytes, the disease mechanisms of SRNS remain poorly understood and no universally safe and effective therapy exists to treat patients with this condition. Recently, genetic research has identified a subgroup of SRNS patients whose kidney pathology is caused by primary coenzyme Q10 (CoQ10) deficiency due to recessive mutations in genes that encode proteins in the CoQ10 biosynthesis pathway. Clinical and preclinical studies show that primary CoQ10 deficiency may be responsive to treatment with CoQ10 supplements bypassing the biosynthesis defects. Coenzyme Q10 is an essential component of the mitochondrial respiratory chain, where it transports electrons from complexes I and II to complex III. Studies in yeast and mammalian model systems have recently identified the molecular functions of the individual CoQ10 biosynthesis complex proteins, validated these findings, and provided an impetus for developing therapeutic compounds to replenish CoQ10 levels in the tissues/organs and thus prevent the destruction of tissues due to mitochondrial OXPHOS deficiencies. In this review, we will summarize the clinical findings of the kidney pathophysiology of primary CoQ10 deficiencies and discuss recent advances in the development of therapies to counter CoQ10 deficiency in tissues.
Collapse
|
18
|
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: 0.5] [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
|
19
|
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: 1.8] [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
|
20
|
Walker MA, Miranda M, Allred A, Mootha VK. On the dynamic and even reversible nature of Leigh syndrome: Lessons from human imaging and mouse models. Curr Opin Neurobiol 2021; 72:80-90. [PMID: 34656053 PMCID: PMC8901530 DOI: 10.1016/j.conb.2021.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/01/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022]
Abstract
Leigh syndrome (LS) is a neurodegenerative disease characterized by bilaterally symmetric brainstem or basal ganglia lesions. More than 80 genes, largely impacting mitochondrial energy metabolism, can underlie LS, and no approved medicines exist. Described 70 years ago, LS was initially diagnosed by the characteristic, necrotic lesions on autopsy. It has been broadly assumed that antemortem neuroimaging abnormalities in these regions correspond to end-stage histopathology. However, clinical observations and animal studies suggest that neuroimaging findings may represent an intermediate state, that is more dynamic than previously appreciated, and even reversible. We review this literature, discuss related conditions that are treatable, and present two new LS cases with radiographic improvement. We review studies in which hypoxia reverses advanced LS in a mouse model. The fluctuating and potentially reversible nature of radiographic LS lesions will be important in clinical trial design. Better understanding of this plasticity could lead to new therapies.
Collapse
Affiliation(s)
- Melissa A Walker
- Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, United States; Broad Institute of Harvard, MIT, United States; Department of Neurology, Massachusetts General Hospital, United States.
| | - Maria Miranda
- Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, United States; Broad Institute of Harvard, MIT, United States
| | - Amanda Allred
- Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, United States
| | - Vamsi K Mootha
- Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, United States; Broad Institute of Harvard, MIT, United States.
| |
Collapse
|
21
|
Minimal mitochondrial respiration is required to prevent cell death by inhibition of mTOR signaling in CoQ-deficient cells. Cell Death Discov 2021; 7:201. [PMID: 34349107 PMCID: PMC8338951 DOI: 10.1038/s41420-021-00591-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/01/2021] [Accepted: 05/23/2021] [Indexed: 01/07/2023] Open
Abstract
Coenzyme Q (CoQ) is a lipid-like mobile electron transporter of the mitochondrial respiratory chain. Patients with partial loss-of-function mutations in the CoQ biosynthesis pathway suffer from partial primary CoQ deficiency (MIM 607426). This leads to mitochondrial dysfunction, which presents like mitochondrial disease syndrome (MDS). In addition, many other conditions, including MDS itself, lead to secondary CoQ deficiency. We sought to identify drugs that can alleviate the consequences of the mitochondrial dysfunction that is associated with CoQ deficiency. Loss of the CoQ-biosynthetic enzyme COQ7 prevents CoQ synthesis but leads to the accumulation of the biosynthetic intermediate demethoxyubiquinone (DMQ). Coq7-knockout mouse embryonic fibroblasts (MEFs) die when rapid ATP generation from glycolysis is prevented. We screened for drugs that could rescue cell death under these conditions. All compounds that were identified inhibit mTOR signaling. In the CoQ-deficient cells, the beneficial action mTOR inhibition appears to be mediated by inhibition of protein translation rather than by stimulation of autophagy. We further studied the Coq7-knockout cells to better determine under which conditions mTOR inhibition could be beneficial. We established that Coq7-knockout cells remain capable of a low level of mitochondrial respiration mediated by DMQ. To obtain more profound mitochondrial dysfunction, we created double-knockout mutant MEFs lacking both Coq7, as well as Pdss2, which is required for sidechain synthesis. These cells make neither CoQ nor DMQ, and their extremely small residual respiration depends on uptake of CoQ from the culture medium. Although these cells are healthy in the presence of sufficient glucose for glycolysis and do not require uridine or pyruvate supplementation, mTOR inhibitors were unable to prevent their death in the absence of sufficient glycolysis. We conclude that, for reasons that remain to be elucidated, the energy-sparing benefits of the inhibition of mTOR signaling require a minimally functional respiratory chain.
Collapse
|
22
|
Krizova J, Hulkova M, Capek V, Mlejnek P, Silhavy J, Tesarova M, Zeman J, Hansikova H. Microarray and qPCR Analysis of Mitochondrial Metabolism Activation during Prenatal and Early Postnatal Development in Rats and Humans with Emphasis on CoQ 10 Biosynthesis. BIOLOGY 2021; 10:418. [PMID: 34066731 PMCID: PMC8150536 DOI: 10.3390/biology10050418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 11/16/2022]
Abstract
At the end of the mammalian intra-uterine foetal development, a rapid switch from glycolytic to oxidative metabolism must proceed. Using microarray techniques, qPCR, enzyme activities and coenzyme Q content measurements, we describe perinatal mitochondrial metabolism acceleration in rat liver and skeletal muscle during the perinatal period and correlate the results with those in humans. Out of 1546 mitochondrial genes, we found significant changes in expression in 1119 and 827 genes in rat liver and skeletal muscle, respectively. The most remarkable expression shift occurred in the rat liver at least two days before birth. Coenzyme Q-based evaluation in both the rat model and human tissues showed the same trend: the total CoQ content is low prenatally, significantly increasing after birth in both the liver and skeletal muscle. We propose that an important regulator of rat coenzyme Q biosynthesis might be COQ8A, an atypical kinase involved in the biosynthesis of coenzyme Q. Our microarray data, a total of 16,557 RefSeq (Entrez) genes, have been deposited in NCBI's Gene Expression Omnibus and are freely available to the broad scientific community. Our microarray data could serve as a suitable background for finding key factors regulating mitochondrial metabolism and the preparation of the foetus for the transition to extra-uterine conditions.
Collapse
Affiliation(s)
- Jana Krizova
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| | - Martina Hulkova
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| | - Vaclav Capek
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| | - Petr Mlejnek
- Department of Genetics of Model Diseases, Institute of Physiology AS CR, v.v.i., Videnska 1083, 142 20 Prague 4, Czech Republic; (P.M.); (J.S.)
| | - Jan Silhavy
- Department of Genetics of Model Diseases, Institute of Physiology AS CR, v.v.i., Videnska 1083, 142 20 Prague 4, Czech Republic; (P.M.); (J.S.)
| | - Marketa Tesarova
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| | - Jiri Zeman
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| | - Hana Hansikova
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| |
Collapse
|
23
|
Alcázar-Fabra M, Rodríguez-Sánchez F, Trevisson E, Brea-Calvo G. Primary Coenzyme Q deficiencies: A literature review and online platform of clinical features to uncover genotype-phenotype correlations. Free Radic Biol Med 2021; 167:141-180. [PMID: 33677064 DOI: 10.1016/j.freeradbiomed.2021.02.046] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/13/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
Primary Coenzyme Q (CoQ) deficiencies are clinically heterogeneous conditions and lack clear genotype-phenotype correlations, complicating diagnosis and prognostic assessment. Here we present a compilation of all the symptoms and patients with primary CoQ deficiency described in the literature so far and analyse the most common clinical manifestations associated with pathogenic variants identified in the different COQ genes. In addition, we identified new associations between the age of onset of symptoms and different pathogenic variants, which could help to a better diagnosis and guided treatment. To make these results useable for clinicians, we created an online platform (https://coenzymeQbiology.github.io/clinic-CoQ-deficiency) about clinical manifestations of primary CoQ deficiency that will be periodically updated to incorporate new information published in the literature. Since CoQ primary deficiency is a rare disease, the available data are still limited, but as new patients are added over time, this tool could become a key resource for a more efficient diagnosis of this pathology.
Collapse
Affiliation(s)
- María Alcázar-Fabra
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA and CIBERER, Instituto de Salud Carlos III, Seville, 41013, Spain
| | | | - Eva Trevisson
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Padova, 35128, Italy; Istituto di Ricerca Pediatrica, Fondazione Città della Speranza, Padova, 35128, Italy.
| | - Gloria Brea-Calvo
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA and CIBERER, Instituto de Salud Carlos III, Seville, 41013, Spain.
| |
Collapse
|
24
|
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: 10.3] [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
|
25
|
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: 1.8] [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
|
26
|
Pitceathly RD, Keshavan N, Rahman J, Rahman S. Moving towards clinical trials for mitochondrial diseases. J Inherit Metab Dis 2021; 44:22-41. [PMID: 32618366 PMCID: PMC8432143 DOI: 10.1002/jimd.12281] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 12/11/2022]
Abstract
Primary mitochondrial diseases represent some of the most common and severe inherited metabolic disorders, affecting ~1 in 4,300 live births. The clinical and molecular diversity typified by mitochondrial diseases has contributed to the lack of licensed disease-modifying therapies available. Management for the majority of patients is primarily supportive. The failure of clinical trials in mitochondrial diseases partly relates to the inefficacy of the compounds studied. However, it is also likely to be a consequence of the significant challenges faced by clinicians and researchers when designing trials for these disorders, which have historically been hampered by a lack of natural history data, biomarkers and outcome measures to detect a treatment effect. Encouragingly, over the past decade there have been significant advances in therapy development for mitochondrial diseases, with many small molecules now transitioning from preclinical to early phase human interventional studies. In this review, we present the treatments and management strategies currently available to people with mitochondrial disease. We evaluate the challenges and potential solutions to trial design and highlight the emerging pharmacological and genetic strategies that are moving from the laboratory to clinical trials for this group of disorders.
Collapse
Affiliation(s)
- Robert D.S. Pitceathly
- Department of Neuromuscular DiseasesUCL Queen Square Institute of Neurology and The National Hospital for Neurology and NeurosurgeryLondonUK
| | - Nandaki Keshavan
- Mitochondrial Research GroupUCL Great Ormond Street Institute of Child HealthLondonUK
- Metabolic UnitGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Joyeeta Rahman
- Mitochondrial Research GroupUCL Great Ormond Street Institute of Child HealthLondonUK
| | - Shamima Rahman
- Mitochondrial Research GroupUCL Great Ormond Street Institute of Child HealthLondonUK
- Metabolic UnitGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| |
Collapse
|
27
|
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: 2.4] [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
|
28
|
Ignatenko O, Nikkanen J, Kononov A, Zamboni N, Ince-Dunn G, Suomalainen A. Mitochondrial spongiotic brain disease: astrocytic stress and harmful rapamycin and ketosis effect. Life Sci Alliance 2020; 3:3/9/e202000797. [PMID: 32737078 PMCID: PMC7409372 DOI: 10.26508/lsa.202000797] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/13/2022] Open
Abstract
Astrocyte-specific mtDNA depletion causes spongiotic encephalopathy, aggravated by ketogenic diet or rapamycin. Astrocytes, but not neurons, drive mitochondrial integrated stress response in the CNS. Mitochondrial DNA (mtDNA) depletion syndrome (MDS) is a group of severe, tissue-specific diseases of childhood with unknown pathogenesis. Brain-specific MDS manifests as devastating spongiotic encephalopathy with no curative therapy. Here, we report cell type–specific stress responses and effects of rapamycin treatment and ketogenic diet (KD) in mice with spongiotic encephalopathy mimicking human MDS, as these interventions were reported to improve some mitochondrial disease signs or symptoms. These mice with astrocyte-specific knockout of Twnk gene encoding replicative mtDNA helicase Twinkle (TwKOastro) show wide-spread cell-autonomous astrocyte activation and mitochondrial integrated stress response (ISRmt) induction with major metabolic remodeling of the brain. Mice with neuronal-specific TwKO show no ISRmt. Both KD and rapamycin lead to rapid deterioration and weight loss of TwKOastro and premature trial termination. Although rapamycin had no robust effects on TwKOastro brain pathology, KD exacerbated spongiosis, gliosis, and ISRmt. Our evidence emphasizes that mitochondrial disease treatments and stress responses are tissue- and disease specific. Furthermore, rapamycin and KD are deleterious in MDS-linked spongiotic encephalopathy, pointing to a crucial role of diet and metabolism for mitochondrial disease progression.
Collapse
Affiliation(s)
- Olesia Ignatenko
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Joni Nikkanen
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | | | - Nicola Zamboni
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Gulayse Ince-Dunn
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anu Suomalainen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland .,Neuroscience Center, University of Helsinki, Helsinki, Finland.,HUSlab, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
29
|
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: 0.6] [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
|
30
|
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: 1.6] [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
|
31
|
Dard L, Blanchard W, Hubert C, Lacombe D, Rossignol R. Mitochondrial functions and rare diseases. Mol Aspects Med 2020; 71:100842. [PMID: 32029308 DOI: 10.1016/j.mam.2019.100842] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/26/2019] [Accepted: 12/27/2019] [Indexed: 12/19/2022]
Abstract
Mitochondria are dynamic cellular organelles responsible for a large variety of biochemical processes as energy transduction, REDOX signaling, the biosynthesis of hormones and vitamins, inflammation or cell death execution. Cell biology studies established that 1158 human genes encode proteins localized to mitochondria, as registered in MITOCARTA. Clinical studies showed that a large number of these mitochondrial proteins can be altered in expression and function through genetic, epigenetic or biochemical mechanisms including the interaction with environmental toxics or iatrogenic medicine. As a result, pathogenic mitochondrial genetic and functional defects participate to the onset and the progression of a growing number of rare diseases. In this review we provide an exhaustive survey of the biochemical, genetic and clinical studies that demonstrated the implication of mitochondrial dysfunction in human rare diseases. We discuss the striking diversity of the symptoms caused by mitochondrial dysfunction and the strategies proposed for mitochondrial therapy, including a survey of ongoing clinical trials.
Collapse
Affiliation(s)
- L Dard
- Bordeaux University, 33000, Bordeaux, France; INSERM U1211, 33000, Bordeaux, France; CELLOMET, CGFB-146 Rue Léo Saignat, Bordeaux, France
| | - W Blanchard
- Bordeaux University, 33000, Bordeaux, France; INSERM U1211, 33000, Bordeaux, France; CELLOMET, CGFB-146 Rue Léo Saignat, Bordeaux, France
| | - C Hubert
- Bordeaux University, 33000, Bordeaux, France; INSERM U1211, 33000, Bordeaux, France
| | - D Lacombe
- Bordeaux University, 33000, Bordeaux, France; INSERM U1211, 33000, Bordeaux, France; CHU de Bordeaux, Service de Génétique Médicale, F-33076, Bordeaux, France
| | - R Rossignol
- Bordeaux University, 33000, Bordeaux, France; INSERM U1211, 33000, Bordeaux, France; CELLOMET, CGFB-146 Rue Léo Saignat, Bordeaux, France.
| |
Collapse
|
32
|
Wang Y, Hekimi S. The Complexity of Making Ubiquinone. Trends Endocrinol Metab 2019; 30:929-943. [PMID: 31601461 DOI: 10.1016/j.tem.2019.08.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 12/15/2022]
Abstract
Ubiquinone (UQ, coenzyme Q) is an essential electron transfer lipid in the mitochondrial respiratory chain. It is a main source of mitochondrial reactive oxygen species (ROS) but also has antioxidant properties. This mix of characteristics is why ubiquinone supplementation is considered a potential therapy for many diseases involving mitochondrial dysfunction. Mutations in the ubiquinone biosynthetic pathway are increasingly being identified in patients. Furthermore, secondary ubiquinone deficiency is a common finding associated with mitochondrial disorders and might exacerbate these conditions. Recent developments have suggested that ubiquinone biosynthesis occurs in discrete domains of the mitochondrial inner membrane close to ER-mitochondria contact sites. This spatial requirement for ubiquinone biosynthesis could be the link between secondary ubiquinone deficiency and mitochondrial dysfunction, which commonly results in loss of mitochondrial structural integrity.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biology, McGill University, Montreal, Canada
| | | |
Collapse
|
33
|
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: 50] [Impact Index Per Article: 8.3] [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
|
34
|
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
|
35
|
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: 26] [Impact Index Per Article: 4.3] [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
|
36
|
An Isoprene Lipid-Binding Protein Promotes Eukaryotic Coenzyme Q Biosynthesis. Mol Cell 2019; 73:763-774.e10. [PMID: 30661980 DOI: 10.1016/j.molcel.2018.11.033] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/16/2018] [Accepted: 11/27/2018] [Indexed: 02/06/2023]
Abstract
The biosynthesis of coenzyme Q presents a paradigm for how cells surmount hydrophobic barriers in lipid biology. In eukaryotes, CoQ precursors-among nature's most hydrophobic molecules-must somehow be presented to a series of enzymes peripherally associated with the mitochondrial inner membrane. Here, we reveal that this process relies on custom lipid-binding properties of COQ9. We show that COQ9 repurposes the bacterial TetR fold to bind aromatic isoprenes with high specificity, including CoQ intermediates that likely reside entirely within the bilayer. We reveal a process by which COQ9 associates with cardiolipin-rich membranes and warps the membrane surface to access this cargo. Finally, we identify a molecular interface between COQ9 and the hydroxylase COQ7, motivating a model whereby COQ9 presents intermediates directly to CoQ enzymes. Overall, our results provide a mechanism for how a lipid-binding protein might access, select, and deliver specific cargo from a membrane to promote biosynthesis.
Collapse
|
37
|
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: 33] [Impact Index Per Article: 5.5] [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
|
38
|
Zhou Z, Austin GL, Young LEA, Johnson LA, Sun R. Mitochondrial Metabolism in Major Neurological Diseases. Cells 2018; 7:E229. [PMID: 30477120 PMCID: PMC6316877 DOI: 10.3390/cells7120229] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 01/18/2023] Open
Abstract
Mitochondria are bilayer sub-cellular organelles that are an integral part of normal cellular physiology. They are responsible for producing the majority of a cell's ATP, thus supplying energy for a variety of key cellular processes, especially in the brain. Although energy production is a key aspect of mitochondrial metabolism, its role extends far beyond energy production to cell signaling and epigenetic regulation⁻functions that contribute to cellular proliferation, differentiation, apoptosis, migration, and autophagy. Recent research on neurological disorders suggest a major metabolic component in disease pathophysiology, and mitochondria have been shown to be in the center of metabolic dysregulation and possibly disease manifestation. This review will discuss the basic functions of mitochondria and how alterations in mitochondrial activity lead to neurological disease progression.
Collapse
Affiliation(s)
- Zhengqiu Zhou
- Molecular & Cellular Biochemistry Department, University of Kentucky, Lexington, KY 40536, USA.
| | - Grant L Austin
- Molecular & Cellular Biochemistry Department, University of Kentucky, Lexington, KY 40536, USA.
| | - Lyndsay E A Young
- Molecular & Cellular Biochemistry Department, University of Kentucky, Lexington, KY 40536, USA.
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA.
| | - Ramon Sun
- Molecular & Cellular Biochemistry Department, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
39
|
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: 0.9] [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
|
40
|
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: 4.1] [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
|
41
|
Smith AC, Ito Y, Ahmed A, Schwartzentruber JA, Beaulieu CL, Aberg E, Majewski J, Bulman DE, Horsting-Wethly K, Koning DVD, Rodenburg RJ, Boycott KM, Penney LS. A family segregating lethal neonatal coenzyme Q 10 deficiency caused by mutations in COQ9. J Inherit Metab Dis 2018; 41:719-729. [PMID: 29560582 DOI: 10.1007/s10545-017-0122-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 11/12/2017] [Accepted: 11/30/2017] [Indexed: 11/27/2022]
Abstract
Primary CoQ10 deficiency is a clinically and genetically heterogeneous, autosomal recessive disorder resulting from mutations in genes involved in the synthesis of coenzyme Q10 (CoQ10). To date, mutations in nine proteins required for the biosynthesis of CoQ10 cause CoQ10 deficiency with varying clinical presentations. In 2009 the first patient with mutations in COQ9 was reported in an infant with a neonatal-onset, primary CoQ10 deficiency with multi-system disease. Here we describe four siblings with a previously undiagnosed lethal disorder characterized by oligohydramnios and intrauterine growth restriction, variable cardiomyopathy, anemia, and renal anomalies. The first and third pregnancy resulted in live born babies with abnormal tone who developed severe, treatment unresponsive lactic acidosis after birth and died hours later. Autopsy on one of the siblings demonstrated brain changes suggestive of the subacute necrotizing encephalopathy of Leigh disease. Whole-exome sequencing (WES) revealed the siblings shared compound heterozygous mutations in the COQ9 gene with both variants predicted to affect splicing. RT-PCR on RNA from patient fibroblasts revealed that the c.521 + 2 T > C variant resulted in splicing out of exons 4-5 and the c.711 + 3G > C variant spliced out exon 6, resulting in undetectable levels of COQ9 protein in patient fibroblasts. The biochemical profile of patient fibroblasts demonstrated a drastic reduction in CoQ10 levels. An additional peak on the chromatogram may represent accumulation of demethoxy coenzyme Q (DMQ), which was shown previously to accumulate as a result of a defect in COQ9. This family expands our understanding of this rare metabolic disease and highlights the prenatal onset, clinical variability, severity, and biochemical profile associated with COQ9-related CoQ10 deficiencies.
Collapse
Affiliation(s)
- Amanda C Smith
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Yoko Ito
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Afsana Ahmed
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Jeremy A Schwartzentruber
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Chandree L Beaulieu
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Erika Aberg
- Maritime Medical Genetics Service, IWK Health Centre, 5850 University Avenue, P.O. Box 9700, Halifax, NS, B3K 6R8, Canada
| | - Jacek Majewski
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Dennis E Bulman
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Karina Horsting-Wethly
- Radboud Center for Mitochondrial Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Diana Vermunt-de Koning
- Radboud Center for Mitochondrial Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Richard J Rodenburg
- Radboud Center for Mitochondrial Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kym M Boycott
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Lynette S Penney
- Maritime Medical Genetics Service, IWK Health Centre, 5850 University Avenue, P.O. Box 9700, Halifax, NS, B3K 6R8, Canada.
- Department of Pediatrics, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
42
|
Diogo CV, Yambire KF, Fernández Mosquera L, Branco F T, Raimundo N. Mitochondrial adventures at the organelle society. Biochem Biophys Res Commun 2018; 500:87-93. [PMID: 28456629 PMCID: PMC5930832 DOI: 10.1016/j.bbrc.2017.04.124] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 04/23/2017] [Indexed: 01/16/2023]
Abstract
Mitochondria are constantly communicating with the rest of the cell. Defects in mitochondria underlie severe pathologies, whose mechanisms remain poorly understood. It is becoming increasingly evident that mitochondrial malfunction resonates in other organelles, perturbing their function and their biogenesis. In this manuscript, we review the current knowledge on the cross-talk between mitochondria and other organelles, particularly lysosomes, peroxisomes and the endoplasmic reticulum. Several organelle interactions are mediated by transcriptional programs, and other signaling mechanisms are likely mediating organelle dysfunction downstream of mitochondrial impairments. Many of these organelle crosstalk pathways are likely to have a role in pathological processes.
Collapse
Affiliation(s)
- Cátia V Diogo
- Universitätsmedizin Göttingen, Institute fur Zellbiochemie, Humboldtallee 23, room 01.423, 37073 Göttingen, Germany
| | - King Faisal Yambire
- Universitätsmedizin Göttingen, Institute fur Zellbiochemie, Humboldtallee 23, room 01.423, 37073 Göttingen, Germany; International Max-Planck Research School in Neuroscience, Göttingen, Germany
| | - Lorena Fernández Mosquera
- Universitätsmedizin Göttingen, Institute fur Zellbiochemie, Humboldtallee 23, room 01.423, 37073 Göttingen, Germany
| | - Tiago Branco F
- Universitätsmedizin Göttingen, Institute fur Zellbiochemie, Humboldtallee 23, room 01.423, 37073 Göttingen, Germany
| | - Nuno Raimundo
- Universitätsmedizin Göttingen, Institute fur Zellbiochemie, Humboldtallee 23, room 01.423, 37073 Göttingen, Germany.
| |
Collapse
|
43
|
Estimating the occurrence of primary ubiquinone deficiency by analysis of large-scale sequencing data. Sci Rep 2017; 7:17744. [PMID: 29255295 PMCID: PMC5735152 DOI: 10.1038/s41598-017-17564-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 11/28/2017] [Indexed: 12/19/2022] Open
Abstract
Primary ubiquinone (UQ) deficiency is an important subset of mitochondrial disease that is caused by mutations in UQ biosynthesis genes. To guide therapeutic efforts we sought to estimate the number of individuals who are born with pathogenic variants likely to cause this disorder. We used the NCBI ClinVar database and literature reviews to identify pathogenic genetic variants that have been shown to cause primary UQ deficiency, and used the gnomAD database of full genome or exome sequences to estimate the frequency of both homozygous and compound heterozygotes within seven genetically-defined populations. We used known population sizes to estimate the number of afflicted individuals in these populations and in the mixed population of the USA. We then performed the same analysis on predicted pathogenic loss-of-function and missense variants that we identified in gnomAD. When including only known pathogenic variants, our analysis predicts 1,665 affected individuals worldwide and 192 in the USA. Adding predicted pathogenic variants, our estimate grows to 123,789 worldwide and 1,462 in the USA. This analysis predicts that there are many undiagnosed cases of primary UQ deficiency, and that a large proportion of these will be in developing regions of the world.
Collapse
|
44
|
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: 6.6] [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
|
45
|
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: 220] [Impact Index Per Article: 27.5] [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
|
46
|
Navas-Enamorado I, Bernier M, Brea-Calvo G, de Cabo R. Influence of anaerobic and aerobic exercise on age-related pathways in skeletal muscle. Ageing Res Rev 2017; 37:39-52. [PMID: 28487241 PMCID: PMC5549001 DOI: 10.1016/j.arr.2017.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 04/18/2017] [Accepted: 04/28/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Ignacio Navas-Enamorado
- Translational Gerontology Branch, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Gloria Brea-Calvo
- Centro Andaluz de Biología del Desarrollo and CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide-CSIC-JA, Sevilla 41013, Spain
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA.
| |
Collapse
|
47
|
Quinzii CM, Luna-Sanchez M, Ziosi M, Hidalgo-Gutierrez A, Kleiner G, Lopez LC. The Role of Sulfide Oxidation Impairment in the Pathogenesis of Primary CoQ Deficiency. Front Physiol 2017; 8:525. [PMID: 28790927 PMCID: PMC5525000 DOI: 10.3389/fphys.2017.00525] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/07/2017] [Indexed: 11/23/2022] Open
Abstract
Coenzyme Q (CoQ) is a lipid present in all cell membranes. One of the multiple metabolic functions of CoQ is to transport electrons in the reaction catalyzed by sulfide:quinone oxidoreductase (SQOR), the first enzyme of the oxidation pathway of sulfides (hydrogen sulfide, H2S). Early evidence of a defect in the metabolism of H2S in primary CoQ deficiency came from yeast studies in Schizosaccharomyces pombe strains defective for dps1 and ppt1 (homologs of PDSS1 and COQ2, respectively), which have H2S accumulation. Our recent studies in human skin fibroblasts and in murine models of primary CoQ deficiency show that, also in mammals, decreased CoQ levels cause impairment of H2S oxidation. Patient fibroblasts carrying different mutations in genes encoding proteins involved in CoQ biosynthesis show reduced SQOR activity and protein levels proportional to the levels of CoQ. In Pdss2kd/kd mice, kidney, the only organ clinically affected, shows reduced SQOR levels and downstream enzymes, accumulation of H2S, and glutathione depletion. Pdss2kd/kd mice have also low levels of thiosulfate in plasma and urine, and increased C4–C6 acylcarnitines in blood, due to inhibition of short-chain acyl-CoA dehydrogenase. Also in Coq9R239X mice, the symptomatic organ, cerebrum, shows accumulation of H2S, reduced SQOR, increase in thiosulfate sulfurtransferase and sulfite oxidase, and reduction in the levels of glutathione and glutathione enzymes, leading to alteration of the biosynthetic pathways of glutamate, serotonin, and catecholamines. Coq9R239X mice have also reduced blood pressure, possible consequence of H2S-induced vasorelaxation. Since liver is not clinically affected in Pdss2 and Coq9 mutant mice, the effects of the impairment of H2S oxidation in this organ were not investigated, despite its critical role in metabolism. In conclusion, in vitro and in vivo studies of CoQ deficient models provide evidence of tissue-specific H2S oxidation impairment, an additional pathomechanism that should be considered in the understanding and treatment of primary CoQ deficiency.
Collapse
Affiliation(s)
- Catarina M Quinzii
- Department of Neurology, Columbia University Medical CenterNew York, NY, United States
| | - Marta Luna-Sanchez
- Department of Physiology, Faculty of Medicine, University of GranadaGranada, Spain.,MRC Mitochondrial Biology UnitCambridge, United Kingdom
| | - Marcello Ziosi
- Department of Neurology, Columbia University Medical CenterNew York, NY, United States
| | | | - Giulio Kleiner
- Department of Neurology, Columbia University Medical CenterNew York, NY, United States
| | - Luis C Lopez
- Department of Physiology, Faculty of Medicine, University of GranadaGranada, Spain
| |
Collapse
|
48
|
He CH, Black DS, Allan CM, Meunier B, Rahman S, Clarke CF. Human COQ9 Rescues a coq9 Yeast Mutant by Enhancing Coenzyme Q Biosynthesis from 4-Hydroxybenzoic Acid and Stabilizing the CoQ-Synthome. Front Physiol 2017; 8:463. [PMID: 28736527 PMCID: PMC5500610 DOI: 10.3389/fphys.2017.00463] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/16/2017] [Indexed: 11/18/2022] Open
Abstract
Coq9 is required for the stability of a mitochondrial multi-subunit complex, termed the CoQ-synthome, and the deamination step of Q intermediates that derive from para-aminobenzoic acid (pABA) in yeast. In human, mutations in the COQ9 gene cause neonatal-onset primary Q10 deficiency. In this study, we determined whether expression of human COQ9 could complement yeast coq9 point or null mutants. We found that expression of human COQ9 rescues the growth of the temperature-sensitive yeast mutant, coq9-ts19, on a non-fermentable carbon source and increases the content of Q6, by enhancing Q biosynthesis from 4-hydroxybenzoic acid (4HB). To study the mechanism for the rescue by human COQ9, we determined the steady-state levels of yeast Coq polypeptides in the mitochondria of the temperature-sensitive yeast coq9 mutant expressing human COQ9. We show that the expression of human COQ9 significantly increased steady-state levels of yeast Coq4, Coq6, Coq7, and Coq9 at permissive temperature. Human COQ9 polypeptide levels persisted at non-permissive temperature. A small amount of the human COQ9 co-purified with tagged Coq6, Coq6-CNAP, indicating that human COQ9 interacts with the yeast Q-biosynthetic complex. These findings suggest that human COQ9 rescues the yeast coq9 temperature-sensitive mutant by stabilizing the CoQ-synthome and increasing Q biosynthesis from 4HB. This finding provides a powerful approach to studying the function of human COQ9 using yeast as a model.
Collapse
Affiliation(s)
- Cuiwen H He
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, University of California, Los AngelesLos Angeles, CA, United States
| | - Dylan S Black
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, University of California, Los AngelesLos Angeles, CA, United States
| | - Christopher M Allan
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, University of California, Los AngelesLos Angeles, CA, United States
| | - Brigitte Meunier
- Institut de Biologie Intégrative de la Cellule, CEA, Centre National de la Recherche Scientifique, UPSud, Paris-Saclay UniversityGif-sur-Yvette, France
| | - Shamima Rahman
- Metabolic Unit, Great Ormond Street Hospital for Children NHS Foundation TrustLondon, United Kingdom.,Mitochondrial Research Group, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child HealthLondon, United Kingdom
| | - Catherine F Clarke
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, University of California, Los AngelesLos Angeles, CA, United States
| |
Collapse
|
49
|
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: 37] [Impact Index Per Article: 4.6] [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
|
50
|
Lindström R, Lindholm P, Palgi M, Saarma M, Heino TI. In vivo screening reveals interactions between Drosophila Manf and genes involved in the mitochondria and the ubiquinone synthesis pathway. BMC Genet 2017; 18:52. [PMID: 28578657 PMCID: PMC5455201 DOI: 10.1186/s12863-017-0509-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 05/08/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Mesencephalic Astrocyte-derived Neurotrophic Factor (MANF) and Cerebral Dopamine Neurotrophic Factor (CDNF) form an evolutionarily conserved family of neurotrophic factors. Orthologues for MANF/CDNF are the only neurotrophic factors as yet identified in invertebrates with conserved amino acid sequence. Previous studies indicate that mammalian MANF and CDNF support and protect brain dopaminergic system in non-cell-autonomous manner. However, MANF has also been shown to function intracellularly in the endoplasmic reticulum. To date, the knowledge on the interacting partners of MANF/CDNF and signaling pathways they activate is rudimentary. Here, we have employed the Drosophila genetics to screen for potential interaction partners of Drosophila Manf (DmManf) in vivo. RESULTS We first show that DmManf plays a role in the development of Drosophila wing. We exploited this function by using Drosophila UAS-RNAi lines and discovered novel genetic interactions of DmManf with genes known to function in the mitochondria. We also found evidence of an interaction between DmManf and the Drosophila homologue encoding Ku70, the closest structural homologue of SAP domain of mammalian MANF. CONCLUSIONS In addition to the previously known functions of MANF/CDNF protein family, DmManf also interacts with mitochondria-related genes. Our data supports the functional importance of these evolutionarily significant proteins and provides new insights for the future studies.
Collapse
Affiliation(s)
- Riitta Lindström
- Department of Biosciences, University of Helsinki, FI-00014 Helsinki, Finland
- Current affiliation: Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Päivi Lindholm
- Institute of Biotechnology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Mari Palgi
- Department of Chemistry and Biotechnology, Tallinn University of Technology, EE-12618 Tallinn, Estonia
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Tapio I. Heino
- Department of Biosciences, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|