1
|
Budzinska A, Galganski L, Wojcicki K, Jarmuszkiewicz W. Adaptation of mitochondrial bioenergetics to coenzyme Q deficiency in human endothelial cells after chronic exposure to bisphosphonates. Sci Rep 2025; 15:17734. [PMID: 40404831 DOI: 10.1038/s41598-025-02710-8] [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: 01/22/2025] [Accepted: 05/15/2025] [Indexed: 05/24/2025] Open
Abstract
Nitrogen-containing bisphosphonates (N-BPs), widely used in bone disease therapy, inhibit the mevalonate pathway, which affects coenzyme Q (CoQ) biosynthesis and may compromise mitochondrial function, particularly in endothelial cells where oxidative stress and mitochondrial dysfunction contribute to cardiovascular disease. This study examined the effects of chronic six-day exposure of human endothelial cells to N-BPs on mitochondrial bioenergetic functions, focusing on drug-induced mitochondrial CoQ (mtCoQ) deficiency. Compared with the mitochondria of control cells, those of endothelial cells treated with 5 µM alendronate or 1 µM zoledronate presented a significant 45-50% decrease in total mtCoQ pool, loss of reduced (mtCoQH2) antioxidant mtCoQ pool, and elevated mitochondrial antioxidant protein superoxide dismutase 2 (SOD2) and uncoupling protein 2 (UCP2) levels. Exposing endothelial cells to N-BPs also led to an overall reduction in mitochondrial substrate oxidation, except for increased fatty acid oxidation. Additionally, the mitochondria of N-BP-treated endothelial cells presented decreased respiratory rates, membrane potential, and ATP synthesis efficiency, and increased H2O2 production resulting from increased mtCoQ reduction during the oxidation of complex I (CI) and CII substrates. N-BP-induced mtCoQ deficiency also resulted in rearranged respiratory chain supercomplexes, particularly downregulation of the III2 + IV supercomplex, and decreased CII, CIII, and CV protein levels and activities. Despite the N-BP-induced decrease in a-heme levels, maximal CIV activity remained unaffected in endothelial mitochondria. These findings highlight the role of N-BPs in disrupting mtCoQ redox homeostasis and associated bioenergetic functions in endothelial mitochondria.
Collapse
Affiliation(s)
- Adrianna Budzinska
- Mitochondrial Biochemistry Research Group, Laboratory of Mitochondrial Biochemistry, Faculty of Biology, Collegium Biologicum, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, 61‑614, Poznan, Poland
| | - Lukasz Galganski
- Mitochondrial Biochemistry Research Group, Laboratory of Mitochondrial Biochemistry, Faculty of Biology, Collegium Biologicum, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, 61‑614, Poznan, Poland.
| | - Krzysztof Wojcicki
- Mitochondrial Biochemistry Research Group, Laboratory of Mitochondrial Biochemistry, Faculty of Biology, Collegium Biologicum, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, 61‑614, Poznan, Poland
| | - Wieslawa Jarmuszkiewicz
- Mitochondrial Biochemistry Research Group, Laboratory of Mitochondrial Biochemistry, Faculty of Biology, Collegium Biologicum, Adam Mickiewicz University, Uniwersytetu Poznanskiego 6, 61‑614, Poznan, Poland.
| |
Collapse
|
2
|
Ji D, Mylvaganam S, Ravi Chander P, Tarnopolsky M, Murphy K, Carlen P. Mitochondria and oxidative stress in epilepsy: advances in antioxidant therapy. Front Pharmacol 2025; 15:1505867. [PMID: 40177125 PMCID: PMC11961640 DOI: 10.3389/fphar.2024.1505867] [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: 10/03/2024] [Accepted: 12/26/2024] [Indexed: 04/05/2025] Open
Abstract
Epilepsy, affecting approximately 50 million individuals worldwide, is a neurological disorder characterized by recurrent seizures. Mitochondrial dysfunction and oxidative stress are critical factors in its pathophysiology, leading to neuronal hyperexcitability and cell death. Because of the multiple mitochondrial pathways that can be involved in epilepsy and mitochondrial dysfunction, it is optimal to treat epilepsy with multiple antioxidants in combination. Recent advancements highlight the potential of antioxidant therapy as a novel treatment strategy. This approach involves tailoring antioxidant interventions-such as melatonin, idebenone, and plant-derived compounds-based on individual mitochondrial health, including mitochondrial DNA mutations and haplogroups that influence oxidative stress susceptibility and treatment response. By combining antioxidants that target multiple pathways, reducing oxidative stress, modulating neurotransmitter systems, and attenuating neuroinflammation, synergistic effects can be achieved, enhancing therapeutic efficacy beyond that of a single antioxidant on its own. Future directions include conducting clinical trials to evaluate these combination therapies, and to translate preclinical successes into effective clinical interventions. Targeting oxidative stress and mitochondrial dysfunction through combination antioxidant therapy represents a promising adjunctive strategy to modify disease progression and improve outcomes for individuals living with epilepsy.
Collapse
Affiliation(s)
- Delphine Ji
- Krembil Research Institute, Toronto, ON, Canada
- Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
| | | | | | - Mark Tarnopolsky
- Department of Pediatrics, McMaster Children’s Hospital, Hamilton, ON, Canada
| | | | - Peter Carlen
- Krembil Research Institute, Toronto, ON, Canada
- Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
- Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Ercanbrack WS, Dungan A, Gaul E, Ramirez M, J. DelVecchio A, Grass C, Wingert RA. Frataxin is essential for zebrafish embryogenesis and pronephros formation. Front Cell Dev Biol 2024; 12:1496244. [PMID: 39723241 PMCID: PMC11669007 DOI: 10.3389/fcell.2024.1496244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 10/29/2024] [Indexed: 12/28/2024] Open
Abstract
Background and objectives Friedreich's Ataxia (FRDA) is a genetic disease that affects a variety of different tissues. The disease is caused by a mutation in the frataxin gene (FXN) which is important for the synthesis of iron-sulfur clusters. The primary pathologies of FRDA are loss of motor control and cardiomyopathy. These occur due to the accumulation of reactive oxygen species (ROS) in the brain and the heart due to their high metabolic rates. Our research aims to understand how developmental processes and the kidney are impacted by a deficiency of FXN. Methods We utilized an antisense oligomer, or morpholino, to knockdown the frataxin gene (fxn) in zebrafish embryos. Knockdown was confirmed via RT-PCR, gel electrophoresis, and Sanger sequencing. To investigate phenotypes, we utilized several staining techniques including whole mount in situ hybridization, Alcian blue, and acridine orange, as well as dextran-FITC clearance assays. Results fxn deficient animals displayed otolith malformations, edema, and reduced survival. Alcian blue staining revealed craniofacial defects in fxn deficient animals, and gene expression studies showed that the pronephros, or embryonic kidney, had several morphological defects. We investigated the function of the pronephros through clearance assays and found that the renal function is disrupted in fxn deficient animals in addition to proximal tubule endocytosis. Utilizing acridine orange staining, we found that cell death is a partial contributor to these phenotypes. Discussion and conclusion This work provides new insights about how fxn deficiency impacts development and kidney morphogenesis. Additionally, this work establishes an additional model system to study FRDA.
Collapse
Affiliation(s)
- Wesley S. Ercanbrack
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | | | | | | | | | | | - Rebecca A. Wingert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
4
|
Kim MB, Lee J, Lee JY. Targeting Mitochondrial Dysfunction for the Prevention and Treatment of Metabolic Disease by Bioactive Food Components. J Lipid Atheroscler 2024; 13:306-327. [PMID: 39355406 PMCID: PMC11439752 DOI: 10.12997/jla.2024.13.3.306] [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: 03/14/2024] [Revised: 04/21/2024] [Accepted: 05/13/2024] [Indexed: 10/03/2024] Open
Abstract
Dysfunctional mitochondria have been linked to the pathogenesis of obesity-associated metabolic diseases. Excessive energy intake impairs mitochondrial biogenesis and function, decreasing adenosine-5'-triphosphate production and negatively impacting metabolically active tissues such as adipose tissue, skeletal muscle, and the liver. Compromised mitochondrial function disturbs lipid metabolism and increases reactive oxygen species production in these tissues, contributing to the development of insulin resistance, type 2 diabetes, and non-alcoholic fatty liver disease. Recent studies have demonstrated the therapeutic potential of bioactive food components, such as resveratrol, quercetin, coenzyme Q10, curcumin, and astaxanthin, by enhancing mitochondrial function. This review provides an overview of the current understanding of how these bioactive compounds ameliorate mitochondrial dysfunction to mitigate obesity-associated metabolic diseases.
Collapse
Affiliation(s)
- Mi-Bo Kim
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
| | - Jaeeun Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
| | - Ji-Young Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
5
|
Dominiak K, Galganski L, Budzinska A, Jarmuszkiewicz W. Coenzyme Q deficiency in endothelial mitochondria caused by hypoxia; remodeling of the respiratory chain and sensitivity to anoxia/reoxygenation. Free Radic Biol Med 2024; 214:158-170. [PMID: 38364943 DOI: 10.1016/j.freeradbiomed.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024]
Abstract
This study examined the effects of hypoxia on coenzyme Q (Q) levels and mitochondrial function in EA. hy926 endothelial cells, shedding light on their responses to changes in oxygen levels. Chronic hypoxia during endothelial cell culture reduced Q synthesis by reducing hydroxy-methylglutaryl-CoA reductase (HMGCR) levels via hypoxia-inducible factor 1α (HIF1α), leading to severe Q deficiency. In endothelial mitochondria, hypoxia led to reorganization of the respiratory chain through upregulation of supercomplexes (I+III2+IV), forming a complete mitochondrial Q (mQ)-mediated electron transfer pathway. Mitochondria of endothelial cells cultured under hypoxic conditions showed reduced respiratory rates and membrane potential, as well as increased production of mitochondrial reactive oxygen species (mROS) as a result of increased mQ reduction levels (mQH2/mQtot). Anoxia/reoxygenation (A/R) in vitro caused impairment of endothelial mitochondria, manifested by reduced maximal respiration, complex III activity, membrane potential, coupling parameters, and increased mQ reduction and mROS production. Weaker A/R-induced changes compared to control mitochondria indicated better tolerance of A/R stress by the mitochondria of hypoxic cells. Moreover, in endothelial mitochondria, hypoxia-induced increases in uncoupling protein 3 (UCP3) and mitochondrial large-conductance Ca2+-activated potassium channel (mitoBKCa) levels and activities appear to have alleviated reoxygenation injury after A/R. These results not only highlight hypoxia-induced changes in mQ redox homeostasis and related mitochondrial function, but also indicate that chronic hypoxia during endothelial cell culture leads to mitochondrial adaptations that help mitochondria better withstand subsequent oxygen fluctuations.
Collapse
Affiliation(s)
- Karolina Dominiak
- Laboratory of Mitochondrial Biochemistry, Department of Bioenergetics, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland.
| | - Lukasz Galganski
- Laboratory of Mitochondrial Biochemistry, Department of Bioenergetics, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland.
| | - Adrianna Budzinska
- Laboratory of Mitochondrial Biochemistry, Department of Bioenergetics, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland.
| | - Wieslawa Jarmuszkiewicz
- Laboratory of Mitochondrial Biochemistry, Department of Bioenergetics, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland.
| |
Collapse
|
6
|
Sun Z, Gao Z, Xiang M, Feng Y, Wang J, Xu J, Wang Y, Liang J. Comprehensive analysis of lactate-related gene profiles and immune characteristics in lupus nephritis. Front Immunol 2024; 15:1329009. [PMID: 38455045 PMCID: PMC10917958 DOI: 10.3389/fimmu.2024.1329009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Objectives The most frequent cause of kidney damage in systemic lupus erythematosus (SLE) is lupus nephritis (LN), which is also a significant risk factor for morbidity and mortality. Lactate metabolism and protein lactylation might be related to the development of LN. However, there is still a lack of relative research to prove the hypothesis. Hence, this study was conducted to screen the lactate-related biomarkers for LN and analyze the underlying mechanism. Methods To identify differentially expressed genes (DEGs) in the training set (GSE32591, GSE127797), we conducted a differential expression analysis (LN samples versus normal samples). Then, module genes were mined using WGCNA concerning LN. The overlapping of DEGs, critical module genes, and lactate-related genes (LRGs) was used to create the lactate-related differentially expressed genes (LR-DEGs). By using a machine-learning algorithm, ROC, and expression levels, biomarkers were discovered. We also carried out an immune infiltration study based on biomarkers and GSEA. Results A sum of 1259 DEGs was obtained between LN and normal groups. Then, 3800 module genes in reference to LN were procured. 19 LR-DEGs were screened out by the intersection of DEGs, key module genes, and LRGs. Moreover, 8 pivotal genes were acquired via two machine-learning algorithms. Subsequently, 3 biomarkers related to lactate metabolism were obtained, including COQ2, COQ4, and NDUFV1. And these three biomarkers were enriched in pathways 'antigen processing and presentation' and 'NOD-like receptor signaling pathway'. We found that Macrophages M0 and T cells regulatory (Tregs) were associated with these three biomarkers as well. Conclusion Overall, the results indicated that lactate-related biomarkers COQ2, COQ4, and NDUFV1 were associated with LN, which laid a theoretical foundation for the diagnosis and treatment of LN.
Collapse
Affiliation(s)
- Zhan Sun
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhanyan Gao
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Mengmeng Xiang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yang Feng
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Wang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Yilun Wang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun Liang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Grayson C, Mailloux RJ. Coenzyme Q 10 and nicotinamide nucleotide transhydrogenase: Sentinels for mitochondrial hydrogen peroxide signaling. Free Radic Biol Med 2023; 208:260-271. [PMID: 37573896 DOI: 10.1016/j.freeradbiomed.2023.08.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/21/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Mitochondria use hydrogen peroxide (H2O2) as a mitokine for cell communication. H2O2 output for signaling depends on its rate of production and degradation, both of which are strongly affected by the redox state of the coenzyme Q10 (CoQ) pool and NADPH availability. Here, we propose the CoQ pool and nicotinamide nucleotide transhydrogenase (NNT) have evolved to be central modalities for mitochondrial H2O2 signaling. Both factors play opposing yet equally important roles in dictating H2O2 availability because they are connected to one another by two central parameters in bioenergetics: electron supply and Δp. The CoQ pool is the central point of convergence for electrons from various dehydrogenases and the electron transport chain (ETC). The increase in Δp creates a significant amount of protonic backpressure on mitochondria to promote H2O2 genesis through CoQ pool reduction. These same factors also drive the activity of NNT, which uses electrons and the Δp to eliminate H2O2. In this way, electron supply and the magnitude of the Δp manifests as a redox connection between the two sentinels, CoQ and NNT, which serve as opposing yet equally important forces required for budgeting H2O2. Taken together, CoQ and NNT are sentinels linked through mitochondrial bioenergetics to manage H2O2 availability for interorganelle and intercellular redox signaling.
Collapse
Affiliation(s)
- Cathryn Grayson
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Ryan J Mailloux
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
8
|
Hong S, Kim S, Kim K, Lee H. Clinical Approaches for Mitochondrial Diseases. Cells 2023; 12:2494. [PMID: 37887337 PMCID: PMC10605124 DOI: 10.3390/cells12202494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
Mitochondria are subcontractors dedicated to energy production within cells. In human mitochondria, almost all mitochondrial proteins originate from the nucleus, except for 13 subunit proteins that make up the crucial system required to perform 'oxidative phosphorylation (OX PHOS)', which are expressed by the mitochondria's self-contained DNA. Mitochondrial DNA (mtDNA) also encodes 2 rRNA and 22 tRNA species. Mitochondrial DNA replicates almost autonomously, independent of the nucleus, and its heredity follows a non-Mendelian pattern, exclusively passing from mother to children. Numerous studies have identified mtDNA mutation-related genetic diseases. The consequences of various types of mtDNA mutations, including insertions, deletions, and single base-pair mutations, are studied to reveal their relationship to mitochondrial diseases. Most mitochondrial diseases exhibit fatal symptoms, leading to ongoing therapeutic research with diverse approaches such as stimulating the defective OXPHOS system, mitochondrial replacement, and allotropic expression of defective enzymes. This review provides detailed information on two topics: (1) mitochondrial diseases caused by mtDNA mutations, and (2) the mechanisms of current treatments for mitochondrial diseases and clinical trials.
Collapse
Affiliation(s)
- Seongho Hong
- Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea;
- Department of Medicine, Korea University College of Medicine, Seoul 02708, Republic of Korea
| | - Sanghun Kim
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea;
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Kyoungmi Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Department of Physiology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hyunji Lee
- Department of Medicine, Korea University College of Medicine, Seoul 02708, Republic of Korea
| |
Collapse
|
9
|
Khanra S, Reddy P, Giménez-Palomo A, Park CHJ, Panizzutti B, McCallum M, Arumugham SS, Umesh S, Debnath M, Das B, Venkatasubramanian G, Ashton M, Turner A, Dean OM, Walder K, Vieta E, Yatham LN, Pacchiarotti I, Reddy YCJ, Goyal N, Kesavan M, Colomer L, Berk M, Kim JH. Metabolic regulation to treat bipolar depression: mechanisms and targeting by trimetazidine. Mol Psychiatry 2023; 28:3231-3242. [PMID: 37386057 PMCID: PMC10618096 DOI: 10.1038/s41380-023-02134-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/14/2023] [Accepted: 06/13/2023] [Indexed: 07/01/2023]
Abstract
Bipolar disorder's core feature is the pathological disturbances in mood, often accompanied by disrupted thinking and behavior. Its complex and heterogeneous etiology implies that a range of inherited and environmental factors are involved. This heterogeneity and poorly understood neurobiology pose significant challenges to existing drug development paradigms, resulting in scarce treatment options, especially for bipolar depression. Therefore, novel approaches are needed to discover new treatment options. In this review, we first highlight the main molecular mechanisms known to be associated with bipolar depression-mitochondrial dysfunction, inflammation and oxidative stress. We then examine the available literature for the effects of trimetazidine in said alterations. Trimetazidine was identified without a priori hypothesis using a gene-expression signature for the effects of a combination of drugs used to treat bipolar disorder and screening a library of off-patent drugs in cultured human neuronal-like cells. Trimetazidine is used to treat angina pectoris for its cytoprotective and metabolic effects (improved glucose utilization for energy production). The preclinical and clinical literature strongly support trimetazidine's potential to treat bipolar depression, having anti-inflammatory and antioxidant properties while normalizing mitochondrial function only when it is compromised. Further, trimetazidine's demonstrated safety and tolerability provide a strong rationale for clinical trials to test its efficacy to treat bipolar depression that could fast-track its repurposing to address such an unmet need as bipolar depression.
Collapse
Affiliation(s)
- Sourav Khanra
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| | - Preethi Reddy
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, India
| | - Anna Giménez-Palomo
- Bipolar and Depressive Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Biomedical Research Networking Center (CIBERSAM), Madrid, Spain
| | - Chun Hui J Park
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Bruna Panizzutti
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Madeleine McCallum
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Shyam Sundar Arumugham
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, India
| | - Shreekantiah Umesh
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| | - Monojit Debnath
- Department of Human Genetics, NIMHANS, Bengaluru, Karnataka, India
| | - Basudeb Das
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| | - Ganesan Venkatasubramanian
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, India
| | - Melanie Ashton
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Alyna Turner
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Olivia M Dean
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Ken Walder
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Eduard Vieta
- Bipolar and Depressive Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Biomedical Research Networking Center (CIBERSAM), Madrid, Spain
| | - Lakshmi N Yatham
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Isabella Pacchiarotti
- Bipolar and Depressive Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Biomedical Research Networking Center (CIBERSAM), Madrid, Spain
| | - Y C Janardhan Reddy
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, India
| | - Nishant Goyal
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| | - Muralidharan Kesavan
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, India
| | - Lluc Colomer
- Bipolar and Depressive Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Biomedical Research Networking Center (CIBERSAM), Madrid, Spain
| | - Michael Berk
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia.
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| | - Jee Hyun Kim
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia.
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
10
|
Yang HL, Huang ST, Lyu ZH, Bhat AA, Vadivalagan C, Yeh YL, Hseu YC. The anti-tumor activities of coenzyme Q0 through ROS-mediated autophagic cell death in human triple-negative breast cells. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
|
11
|
Kornblum C, Lamperti C, Parikh S. Currently available therapies in mitochondrial disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 194:189-206. [PMID: 36813313 DOI: 10.1016/b978-0-12-821751-1.00007-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Mitochondrial diseases are a heterogeneous group of multisystem disorders caused by impaired mitochondrial function. These disorders occur at any age and involve any tissue, typically affecting organs highly dependent on aerobic metabolism. Diagnosis and management are extremely difficult due to various underlying genetic defects and a wide range of clinical symptoms. Preventive care and active surveillance are strategies to try to reduce morbidity and mortality by timely treatment of organ-specific complications. More specific interventional therapies are in early phases of development and no effective treatment or cure currently exists. A variety of dietary supplements have been utilized based on biological logic. For several reasons, few randomized controlled trials have been completed to assess the efficacy of these supplements. The majority of the literature on supplement efficacy represents case reports, retrospective analyses and open-label studies. We briefly review selected supplements that have some degree of clinical research support. In mitochondrial diseases, potential triggers of metabolic decompensation or medications that are potentially toxic to mitochondrial function should be avoided. We shortly summarize current recommendations on safe medication in mitochondrial diseases. Finally, we focus on the frequent and debilitating symptoms of exercise intolerance and fatigue and their management including physical training strategies.
Collapse
Affiliation(s)
- Cornelia Kornblum
- Department of Neurology, Neuromuscular Disease Section, University Hospital Bonn, Bonn, Germany.
| | - Costanza Lamperti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sumit Parikh
- Center for Pediatric Neurosciences, Mitochondrial Medicine & Neurogenetics, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
12
|
Chang CF, Gunawan AL, Liparulo I, Zushin PJH, Bertholet AM, Kirichok Y, Stahl A. CoQ Regulates Brown Adipose Tissue Respiration and Uncoupling Protein 1 Expression. Antioxidants (Basel) 2022; 12:14. [PMID: 36670876 PMCID: PMC9854525 DOI: 10.3390/antiox12010014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Coenzyme Q (CoQ, aka ubiquinone) is a key component of the mitochondrial electron transport chain (ETC) and membrane-incorporated antioxidant. CoQ10 deficiencies encompass a heterogeneous spectrum of clinical phenotypes and can be caused by hereditary mutations in the biosynthesis pathway or result from pharmacological interventions such as HMG-CoA Reductase inhibitors, and statins, which are widely used to treat hypercholesterolemia and prevent cardiovascular disease. How CoQ deficiency affects individual tissues and cell types, particularly mitochondrial-rich ones such as brown adipose tissue (BAT), has remained poorly understood. Here we show that pharmacological and genetic models of BAT CoQ deficiency show altered respiration that can only in part be explained by classical roles of CoQ in the respiration chain. Instead, we found that CoQ strongly impacts brown and beige adipocyte respiration via the regulation of uncoupling protein 1 (UCP1) expression. CoQ deficiency in BAT robustly decreases UCP1 protein levels and uncoupled respiration unexpectedly, resulting in increased inner mitochondrial membrane potential and decreased ADP/ATP ratios. Suppressed UCP1 expression was also observed in a BAT-specific in vivo model of CoQ deficiency and resulted in enhanced cold sensitivity. These findings demonstrate an as yet unappreciated role of CoQ in the transcriptional regulation of key thermogenic genes and functions.
Collapse
Affiliation(s)
- Ching-Fang Chang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Amanda L. Gunawan
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Irene Liparulo
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Peter-James H. Zushin
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Ambre M. Bertholet
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuriy Kirichok
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
13
|
Quantum tunnelling in the context of SARS-CoV-2 infection. Sci Rep 2022; 12:16929. [PMID: 36209224 PMCID: PMC9547378 DOI: 10.1038/s41598-022-21321-1] [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: 02/04/2022] [Accepted: 09/26/2022] [Indexed: 12/29/2022] Open
Abstract
The SARS-CoV-2 pandemic has added new urgency to the study of viral mechanisms of infection. But while vaccines offer a measure of protection against this specific outbreak, a new era of pandemics has been predicted. In addition to this, COVID-19 has drawn attention to post-viral syndromes and the healthcare burden they entail. It seems integral that knowledge of viral mechanisms is increased through as wide a research field as possible. To this end we propose that quantum biology might offer essential new insights into the problem, especially with regards to the important first step of virus-host invasion. Research in quantum biology often centres around energy or charge transfer. While this is predominantly in the context of photosynthesis there has also been some suggestion that cellular receptors such as olfactory or neural receptors might employ vibration assisted electron tunnelling to augment the lock-and-key mechanism. Quantum tunnelling has also been observed in enzyme function. Enzymes are implicated in the invasion of host cells by the SARS-CoV-2 virus. Receptors such as olfactory receptors also appear to be disrupted by COVID-19. Building on these observations we investigate the evidence that quantum tunnelling might be important in the context of infection with SARS-CoV-2. We illustrate this with a simple model relating the vibronic mode of, for example, a viral spike protein to the likelihood of charge transfer in an idealised receptor. Our results show a distinct parameter regime in which the vibronic mode of the spike protein enhances electron transfer. With this in mind, novel therapeutics to prevent SARS-CoV-2 transmission could potentially be identified by their vibrational spectra.
Collapse
|
14
|
Trajectory of the spectral/structural rearrangements for photo-oxidative reaction of neat ketoprofen and its cyclodextrin complex. J INCL PHENOM MACRO 2022. [DOI: 10.1007/s10847-022-01160-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
15
|
Hernández-Pérez OR, Juárez-Navarro KJ, Diaz NF, Padilla-Camberos E, Beltran-Garcia MJ, Cardenas-Castrejon D, Corona-Perez H, Hernández-Jiménez C, Díaz-Martínez NE. Biomolecules resveratrol + coenzyme Q10 recover the cell state of human mesenchymal stem cells after 1-methyl-4-phenylpyridinium-induced damage and improve proliferation and neural differentiation. Front Neurosci 2022; 16:929590. [PMID: 36117620 PMCID: PMC9471188 DOI: 10.3389/fnins.2022.929590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/04/2022] [Indexed: 11/20/2022] Open
Abstract
Neurodegenerative disorders are a critical affection with a high incidence around the world. Currently, there are no effective treatments to solve this problem. However, the application of mesenchymal stem cells (MSCs) and antioxidants in neurodegenerative diseases has shown to be a promising tool due to their multiple therapeutic effects. This work aimed to evaluate the effects of a combination of resveratrol (RSV) and coenzyme Q10 (CoQ10) on the proliferation and differentiation of MSC and the protector effects in induced damage. To characterize the MSCs, we performed flow cytometry, protocols of cellular differentiation, and immunocytochemistry analysis. The impact of RSV + CoQ10 in proliferation was evaluated by supplementing 2.5 and 10 μM of RSV + CoQ10 in a cellular kinetic for 14 days. Cell viability and lactate dehydrogenase levels (LDH) were also analyzed. The protective effect of RSV + CoQ10 was assessed by supplementing the treatment to damaged MSCs by 1-methyl-4-phenylpyridinium (MPP+); cellular viability, LDH, and reactive oxygen species (ROS) were evaluated.. MSCs expressed the surface markers CD44, CD73, CD90, and CD105 and showed multipotential ability. The combination of RSV + CoQ10 increased the proliferation potential and cell viability and decreased LDH levels. In addition, it reverted the effect of MPP+-induced damage in MSCs to enhance cell viability and decrease LDH and ROS. Finally, RSV + CoQ10 promoted the differentiation of neural progenitors. The combination of RSV + CoQ10 represents a potential treatment to improve MSCs capacities and protect against neurodegenerative damage.
Collapse
Affiliation(s)
- Oscar R. Hernández-Pérez
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Karen J. Juárez-Navarro
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Nestor F. Diaz
- Instituto Nacional de Perinatología (INPER), Mexico City, Mexico
| | - Eduardo Padilla-Camberos
- Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Miguel J. Beltran-Garcia
- Departamento de Biotecnológicas y Ambientales, Universidad Autónoma de Guadalajara, Zapopan, Mexico
| | | | | | | | - Néstor E. Díaz-Martínez
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
- *Correspondence: Néstor E. Díaz-Martínez,
| |
Collapse
|
16
|
Staufer T, Schulze ML, Schmutzler O, Körnig C, Welge V, Burkhardt T, Vietzke JP, Vogelsang A, Weise JM, Blatt T, Dabrowski O, Falkenberg G, Brückner D, Sanchez-Cano C, Grüner F. Assessing Cellular Uptake of Exogenous Coenzyme Q 10 into Human Skin Cells by X-ray Fluorescence Imaging. Antioxidants (Basel) 2022; 11:antiox11081532. [PMID: 36009252 PMCID: PMC9405069 DOI: 10.3390/antiox11081532] [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: 06/17/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/23/2022] Open
Abstract
X-ray fluorescence (XRF) imaging is a highly sensitive non-invasive imaging method for detection of small element quantities in objects, from human-sized scales down to single-cell organelles, using various X-ray beam sizes. Our aim was to investigate the cellular uptake and distribution of Q10, a highly conserved coenzyme with antioxidant and bioenergetic properties. Q10 was labeled with iodine (I2-Q10) and individual primary human skin cells were scanned with nano-focused beams. Distribution of I2-Q10 molecules taken up inside the screened individual skin cells was measured, with a clear correlation between individual Q10 uptake and cell size. Experiments revealed that labeling Q10 with iodine causes no artificial side effects as a result of the labeling procedure itself, and thus is a perfect means of investigating bioavailability and distribution of Q10 in cells. In summary, individual cellular Q10 uptake was demonstrated by XRF, opening the path towards Q10 multi-scale tracking for biodistribution studies.
Collapse
Affiliation(s)
- Theresa Staufer
- Universität Hamburg and Center for Free-Electron Laser Science (CFEL), Institute for Experimental Physics, Faculty for Mathematics, Informatics and Natural Sciences, Luruper Chaussee 149, 22761 Hamburg, Germany
- Correspondence:
| | - Mirja L. Schulze
- Research and Development, Beiersdorf AG, Unnastrasse 48, 20245 Hamburg, Germany
| | - Oliver Schmutzler
- Universität Hamburg and Center for Free-Electron Laser Science (CFEL), Institute for Experimental Physics, Faculty for Mathematics, Informatics and Natural Sciences, Luruper Chaussee 149, 22761 Hamburg, Germany
| | - Christian Körnig
- Universität Hamburg and Center for Free-Electron Laser Science (CFEL), Institute for Experimental Physics, Faculty for Mathematics, Informatics and Natural Sciences, Luruper Chaussee 149, 22761 Hamburg, Germany
| | - Vivienne Welge
- Research and Development, Beiersdorf AG, Unnastrasse 48, 20245 Hamburg, Germany
| | - Thorsten Burkhardt
- Research and Development, Beiersdorf AG, Unnastrasse 48, 20245 Hamburg, Germany
| | - Jens-Peter Vietzke
- Research and Development, Beiersdorf AG, Unnastrasse 48, 20245 Hamburg, Germany
| | - Alexandra Vogelsang
- Research and Development, Beiersdorf AG, Unnastrasse 48, 20245 Hamburg, Germany
| | - Julia M. Weise
- Research and Development, Beiersdorf AG, Unnastrasse 48, 20245 Hamburg, Germany
| | - Thomas Blatt
- Research and Development, Beiersdorf AG, Unnastrasse 48, 20245 Hamburg, Germany
| | - Oliver Dabrowski
- Fraunhofer Institute for Applied Polymer Research (IAP), Center for Applied Nanotechnology (CAN), Grindelallee 117, 20146 Hamburg, Germany
| | - Gerald Falkenberg
- Deutsches Elektronen-Synchrotron DESY, Notkestrasse 85, 22607 Hamburg, Germany
| | - Dennis Brückner
- Deutsches Elektronen-Synchrotron DESY, Notkestrasse 85, 22607 Hamburg, Germany
| | - Carlos Sanchez-Cano
- DIPC, Paseo Manuel de Lardizabal 4, 20018 Donostia-San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, Plaza de Euskadi 5, 48009 Bilbao, Spain
| | - Florian Grüner
- Universität Hamburg and Center for Free-Electron Laser Science (CFEL), Institute for Experimental Physics, Faculty for Mathematics, Informatics and Natural Sciences, Luruper Chaussee 149, 22761 Hamburg, Germany
| |
Collapse
|
17
|
Naddafi M, Mehrizi AA, Eghbal MA, Khansari MG, Azarmi Y, Sattari MR, Karaman C, Karimi F, Alizadeh M, Yazdani MN, Hosseinpour P. Reducing the risk of death induced by aluminum phosphide poisoning: The new therapies. CHEMOSPHERE 2022; 294:133800. [PMID: 35101429 DOI: 10.1016/j.chemosphere.2022.133800] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/22/2022] [Accepted: 01/27/2022] [Indexed: 06/14/2023]
Abstract
Numerous people suffer from accidental or deliberate exposure to different pesticides when poisoning with aluminum phosphate (AlP) is increasing in the eastern countries. Aluminum phosphate is a conventional insecticide that quickly reacts with water or the moistures in the atmosphere and produces fatal phosphine gas, which absorbs quickly by the body. Oral consumption or inhalation of AlP leads to excessive reaction of the body such as fatigue, vomiting, fever, palpitation, vasodilatory shock, increasing blood pressure, cardiac dysfunction, pulmonary congestion, shortness of breath, and death. The garlic smell from the patient's mouth or exhale is one of the methods to recognize the positioning. Due to the lack of individual antidotes, several supportive treatments are required. The present study focused on the available and new therapies that help reduce the effect of AlP poisoning and the mortality rate. The therapies are divided into the antioxidant-related agent and the other agents. The impacts of each agent on the experimental cases are reported.
Collapse
Affiliation(s)
- Mastoureh Naddafi
- Pharmacology and Toxicology Department, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbasali Abouei Mehrizi
- School of Physics, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| | - Mohammad Ali Eghbal
- Pharmacology and Toxicology Department, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahmoud Ghazi Khansari
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yadollah Azarmi
- Pharmacology and Toxicology Department, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Sattari
- Pharmacology and Toxicology Department, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ceren Karaman
- Akdeniz University, Department of Electricity and Energy, Antalya, 07070, Turkey.
| | - Fatemeh Karimi
- Department of Chemical Engineering, Quchan University of Technology, Quchan, 9477177870, Iran
| | - Marzieh Alizadeh
- Pharmeceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Laboratory of Basic Sciences, Mohammad Rasul Allah Research Tower, Shiraz University of Medical Sciences, Shiraz, PO Box: 71348-14336, Iran
| | - Mohammad Nima Yazdani
- Laboratory of Basic Sciences, Mohammad Rasul Allah Research Tower, Shiraz University of Medical Sciences, Shiraz, PO Box: 71348-14336, Iran
| | - Parsa Hosseinpour
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
18
|
Kurata K, Hosono K, Takayama M, Katsuno M, Saitsu H, Ogata T, Hotta Y. Retinitis pigmentosa with optic neuropathy and COQ2 mutations: A case report. Am J Ophthalmol Case Rep 2022; 25:101298. [PMID: 35112026 PMCID: PMC8789597 DOI: 10.1016/j.ajoc.2022.101298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 07/28/2021] [Accepted: 01/17/2022] [Indexed: 10/26/2022] Open
|
19
|
Coenzyme Q 0 Inhibits NLRP3 Inflammasome Activation through Mitophagy Induction in LPS/ATP-Stimulated Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4266214. [PMID: 35035661 PMCID: PMC8759827 DOI: 10.1155/2022/4266214] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 12/04/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
Abstract
Coenzyme Q (CoQ) analogs with a variable number of isoprenoid units have exhibited as anti-inflammatory as well as antioxidant molecules. Using novel quinone derivative CoQ0 (2,3-dimethoxy-5-methyl-1,4-benzoquinone, zero side chain isoprenoid), we studied its molecular activities against LPS/ATP-induced inflammation and redox imbalance in murine RAW264.7 macrophages. CoQ0's non- or subcytotoxic concentration suppressed the NLRP3 inflammasome and procaspase-1 activation, followed by downregulation of IL1β expression in LPS/ATP-stimulated RAW264.7 macrophages. Similarly, treatment of CoQ0 led to LC3-I/II accumulation and p62/SQSTM1 activation. An increase in the Beclin-1/Bcl-2 ratio and a decrease in the expression of phosphorylated PI3K/AKT, p70 S6 kinase, and mTOR showed that autophagy was activated. Besides, CoQ0 increased Parkin protein to recruit damaged mitochondria and induced mitophagy in LPS/ATP-stimulated RAW264.7 macrophages. CoQ0 inhibited LPS/ATP-stimulated ROS generation in RAW264.7 macrophages. Notably, when LPS/ATP-stimulated RAW264.7 macrophages were treated with CoQ0, Mito-TEMPO (a mitochondrial ROS inhibitor), or N-acetylcysteine (NAC, a ROS inhibitor), there was a significant reduction of LPS/ATP-stimulated NLRP3 inflammasome activation and IL1β expression. Interestingly, treatment with CoQ0 or Mito-TEMPO, but not NAC, significantly increased LPS/ATP-induced LC3-II accumulation indicating that mitophagy plays a key role in the regulation of CoQ0-inhibited NLRP3 inflammasome activation. Nrf2 knockdown significantly decreased IL1β expression in LPS/ATP-stimulated RAW264.7 macrophages suggesting that CoQ0 inhibited ROS-mediated NLRP3 inflammasome activation and IL1β expression was suppressed due to the Nrf2 activation. Hence, this study showed that CoQ0 might be a promising candidate for the therapeutics of inflammatory disorders due to its effective anti-inflammatory as well as antioxidant properties.
Collapse
|
20
|
Mitochondrial Management of Reactive Oxygen Species. Antioxidants (Basel) 2021; 10:antiox10111824. [PMID: 34829696 PMCID: PMC8614740 DOI: 10.3390/antiox10111824] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 01/10/2023] Open
Abstract
Mitochondria in aerobic eukaryotic cells are both the site of energy production and the formation of harmful species, such as radicals and other reactive oxygen species, known as ROS. They contain an efficient antioxidant system, including low-molecular-mass molecules and enzymes that specialize in removing various types of ROS or repairing the oxidative damage of biological molecules. Under normal conditions, ROS production is low, and mitochondria, which are their primary target, are slightly damaged in a similar way to other cellular compartments, since the ROS released by the mitochondria into the cytosol are negligible. As the mitochondrial generation of ROS increases, they can deactivate components of the respiratory chain and enzymes of the Krebs cycle, and mitochondria release a high amount of ROS that damage cellular structures. More recently, the feature of the mitochondrial antioxidant system, which does not specifically deal with intramitochondrial ROS, was discovered. Indeed, the mitochondrial antioxidant system detoxifies exogenous ROS species at the expense of reducing the equivalents generated in mitochondria. Thus, mitochondria are also a sink of ROS. These observations highlight the importance of the mitochondrial antioxidant system, which should be considered in our understanding of ROS-regulated processes. These processes include cell signaling and the progression of metabolic and neurodegenerative disease.
Collapse
|
21
|
Lee YH, Park JY, Lee H, Song ES, Kuk MU, Joo J, Oh S, Kwon HW, Park JT, Park SC. Targeting Mitochondrial Metabolism as a Strategy to Treat Senescence. Cells 2021; 10:cells10113003. [PMID: 34831224 PMCID: PMC8616445 DOI: 10.3390/cells10113003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
Mitochondria are one of organelles that undergo significant changes associated with senescence. An increase in mitochondrial size is observed in senescent cells, and this increase is ascribed to the accumulation of dysfunctional mitochondria that generate excessive reactive oxygen species (ROS). Such dysfunctional mitochondria are prime targets for ROS-induced damage, which leads to the deterioration of oxidative phosphorylation and increased dependence on glycolysis as an energy source. Based on findings indicating that senescent cells exhibit mitochondrial metabolic alterations, a strategy to induce mitochondrial metabolic reprogramming has been proposed to treat aging and age-related diseases. In this review, we discuss senescence-related mitochondrial changes and consequent mitochondrial metabolic alterations. We assess the significance of mitochondrial metabolic reprogramming for senescence regulation and propose the appropriate control of mitochondrial metabolism to ameliorate senescence. Learning how to regulate mitochondrial metabolism will provide knowledge for the control of aging and age-related pathologies. Further research focusing on mitochondrial metabolic reprogramming will be an important guide for the development of anti-aging therapies, and will provide novel strategies for anti-aging interventions.
Collapse
Affiliation(s)
- Yun Haeng Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.H.L.); (J.Y.P.); (H.L.); (E.S.S.); (M.U.K.); (J.J.)
| | - Ji Yun Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.H.L.); (J.Y.P.); (H.L.); (E.S.S.); (M.U.K.); (J.J.)
| | - Haneur Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.H.L.); (J.Y.P.); (H.L.); (E.S.S.); (M.U.K.); (J.J.)
| | - Eun Seon Song
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.H.L.); (J.Y.P.); (H.L.); (E.S.S.); (M.U.K.); (J.J.)
| | - Myeong Uk Kuk
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.H.L.); (J.Y.P.); (H.L.); (E.S.S.); (M.U.K.); (J.J.)
| | - Junghyun Joo
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.H.L.); (J.Y.P.); (H.L.); (E.S.S.); (M.U.K.); (J.J.)
| | - Sekyung Oh
- Department of Medical Sciences, Catholic Kwandong University College of Medicine, Incheon 22711, Korea;
| | - Hyung Wook Kwon
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.H.L.); (J.Y.P.); (H.L.); (E.S.S.); (M.U.K.); (J.J.)
- Correspondence: (H.W.K.); (J.T.P.); ; (S.C.P.); Tel.: +82-32-835-8090 (H.W.K.); +82-32-835-8841 (J.T.P.); +82-10-5495-9200 (S.C.P.)
| | - Joon Tae Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.H.L.); (J.Y.P.); (H.L.); (E.S.S.); (M.U.K.); (J.J.)
- Correspondence: (H.W.K.); (J.T.P.); ; (S.C.P.); Tel.: +82-32-835-8090 (H.W.K.); +82-32-835-8841 (J.T.P.); +82-10-5495-9200 (S.C.P.)
| | - Sang Chul Park
- The Future Life & Society Research Center, Chonnam National University, Gwangju 61186, Korea
- Correspondence: (H.W.K.); (J.T.P.); ; (S.C.P.); Tel.: +82-32-835-8090 (H.W.K.); +82-32-835-8841 (J.T.P.); +82-10-5495-9200 (S.C.P.)
| |
Collapse
|
22
|
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
|
23
|
Rodríguez-Varela C, Labarta E. Does Coenzyme Q10 Supplementation Improve Human Oocyte Quality? Int J Mol Sci 2021; 22:ijms22179541. [PMID: 34502447 PMCID: PMC8431086 DOI: 10.3390/ijms22179541] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/19/2022] Open
Abstract
Acquiring oocyte competence requires optimal mitochondrial function and adequate ATP levels. In this context, CoQ10 supplementation may improve human oocyte quality and subsequent reproductive performance given its role in ATP synthesis and mitochondrial protection from ROS oxidative damage. In infertility treatments, CoQ10 therapy can be orally supplied to promote a more favorable environment for oocyte development in vivo or by its addition to culture media in an attempt to improve its quality in vitro. Human clinical studies evaluating the impact of CoQ10 on reproductive performance are summarized in this review, although the available data do not clearly prove its ability to improve human oocyte quality. The main objective is to provide readers with a complete overview of this topic's current status as well as the keys for potential future research lines that may help to take this therapy to clinical practice. Indeed, further clinical trials are needed to confirm these results along with molecular studies to evaluate the impact of CoQ10 supplementation on oxidative stress status and mitochondrial function in human gametes.
Collapse
Affiliation(s)
| | - Elena Labarta
- IVI Foundation—IIS La Fe, 46026 Valencia, Spain;
- IVIRMA Valencia, 46015 Valencia, Spain
| |
Collapse
|
24
|
Wincup C, Sawford N, Rahman A. Pathological mechanisms of abnormal iron metabolism and mitochondrial dysfunction in systemic lupus erythematosus. Expert Rev Clin Immunol 2021; 17:957-967. [PMID: 34263712 PMCID: PMC8452144 DOI: 10.1080/1744666x.2021.1953981] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/07/2021] [Indexed: 12/19/2022]
Abstract
Introduction: Systemic lupus erythematosus [SLE] is a chronic, autoimmune condition characterized by the formation of autoantibodies directed against nuclear components and by oxidative stress. Recently, a number of studies have demonstrated the essential role of iron in the immune response and there is growing evidence that abnormal iron homeostasis can occur in the chronic inflammatory state seen in SLE. Not only is iron vital for hematopoiesis, it is also important for a number of other key physiological processes, in particular in maintaining healthy mitochondrial function.Areas covered: In this review, we highlight the latest understanding with regards to how patients with SLE may be at risk of cellular iron depletion as a result of both absolute and functional iron deficiency. Furthermore, we aim to explain the latest evidence of mitochondrial dysfunction in the pathogenesis of the disease.Expert opinion: Growing evidence suggests that both abnormal iron homeostasis and subsequent mitochondrial dysfunction can impair effector immune cell function. Through a greater understanding of these abnormalities, therapeutic options that directly target iron and mitochondria may ultimately represent novel treatment targets that may translate into clinical care of patients with SLE in the near future.
Collapse
Affiliation(s)
- Chris Wincup
- Department of Rheumatology, Division of Medicine, University College London, London, UK
| | - Natalie Sawford
- Department of Rheumatology, Division of Medicine, University College London, London, UK
| | - Anisur Rahman
- Department of Rheumatology, Division of Medicine, University College London, London, UK
| |
Collapse
|
25
|
Gonzalez-Franquesa A, Stocks B, Chubanava S, Hattel HB, Moreno-Justicia R, Peijs L, Treebak JT, Zierath JR, Deshmukh AS. Mass-spectrometry-based proteomics reveals mitochondrial supercomplexome plasticity. Cell Rep 2021; 35:109180. [PMID: 34038727 DOI: 10.1016/j.celrep.2021.109180] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 01/29/2021] [Accepted: 05/04/2021] [Indexed: 11/26/2022] Open
Abstract
Mitochondrial respiratory complex subunits assemble in supercomplexes. Studies of supercomplexes have typically relied upon antibody-based quantification, often limited to a single subunit per respiratory complex. To provide a deeper insight into mitochondrial and supercomplex plasticity, we combine native electrophoresis and mass spectrometry to determine the supercomplexome of skeletal muscle from sedentary and exercise-trained mice. We quantify 422 mitochondrial proteins within 10 supercomplex bands in which we show the debated presence of complexes II and V. Exercise-induced mitochondrial biogenesis results in non-stoichiometric changes in subunits and incorporation into supercomplexes. We uncover the dynamics of supercomplex-related assembly proteins and mtDNA-encoded subunits after exercise. Furthermore, exercise affects the complexing of Lactb, an obesity-associated mitochondrial protein, and ubiquinone biosynthesis proteins. Knockdown of ubiquinone biosynthesis proteins leads to alterations in mitochondrial respiration. Our approach can be applied to broad biological systems. In this instance, comprehensively analyzing respiratory supercomplexes illuminates previously undetectable complexity in mitochondrial plasticity.
Collapse
Affiliation(s)
- Alba Gonzalez-Franquesa
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sabina Chubanava
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Helle B Hattel
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Roger Moreno-Justicia
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Lone Peijs
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 17177, Sweden
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark; Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
26
|
Ullah H, Di Minno A, Santarcangelo C, Khan H, Daglia M. Improvement of Oxidative Stress and Mitochondrial Dysfunction by β-Caryophyllene: A Focus on the Nervous System. Antioxidants (Basel) 2021; 10:546. [PMID: 33915950 PMCID: PMC8066981 DOI: 10.3390/antiox10040546] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/19/2021] [Accepted: 03/28/2021] [Indexed: 02/05/2023] Open
Abstract
Mitochondrial dysfunction results in a series of defective cellular events, including decreased adenosine triphosphate (ATP) production, enhanced reactive oxygen species (ROS) output, and altered proteastasis and cellular quality control. An enhanced output of ROS may damage mitochondrial components, such as mitochondrial DNA and elements of the electron transport chain, resulting in the loss of proper electrochemical gradient across the mitochondrial inner membrane and an ensuing shutdown of mitochondrial energy production. Neurons have an increased demand for ATP and oxygen, and thus are more prone to damage induced by mitochondrial dysfunction. Mitochondrial dysfunction, damaged electron transport chains, altered membrane permeability and Ca2+ homeostasis, and impaired mitochondrial defense systems induced by oxidative stress, are pathological changes involved in neurodegenerative disorders. A growing body of evidence suggests that the use of antioxidants could stabilize mitochondria and thus may be suitable for preventing neuronal loss. Numerous natural products exhibit the potential to counter oxidative stress and mitochondrial dysfunction; however, science is still looking for a breakthrough in the treatment of neurodegenerative disorders. β-caryophyllene is a bicyclic sesquiterpene, and an active principle of essential oils derived from a large number of spices and food plants. As a selective cannabinoid receptor 2 (CB2) agonist, several studies have reported it as possessing numerous pharmacological activities such as antibacterial (e.g., Helicobacter pylori), antioxidant, anti-inflammatory, analgesic (e.g., neuropathic pain), anti-neurodegenerative and anticancer properties. The present review mainly focuses on the potential of β-caryophyllene in reducing oxidative stress and mitochondrial dysfunction, and its possible links with neuroprotection.
Collapse
Affiliation(s)
- Hammad Ullah
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (H.U.); (A.D.M.); (C.S.)
| | - Alessandro Di Minno
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (H.U.); (A.D.M.); (C.S.)
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy
| | - Cristina Santarcangelo
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (H.U.); (A.D.M.); (C.S.)
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan; or
| | - Maria Daglia
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (H.U.); (A.D.M.); (C.S.)
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
27
|
A Theacrine-Based Supplement Increases Cellular NAD + Levels and Affects Biomarkers Related to Sirtuin Activity in C2C12 Muscle Cells In Vitro. Nutrients 2020; 12:nu12123727. [PMID: 33287129 PMCID: PMC7761648 DOI: 10.3390/nu12123727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/21/2022] Open
Abstract
There is evidence in rodents to suggest that theacrine-based supplements modulate tissue sirtuin activity as well as other biological processes associated with aging. Herein, we examined if a theacrine-based supplement (termed NAD3) altered sirtuin activity in vitro while also affecting markers of mitochondrial biogenesis. The murine C2C12 myoblast cell line was used for experimentation. Following 7 days of differentiation, myotubes were treated with 0.45 mg/mL of NAD3 (containing ~2 mM theacrine) for 3 and 24 h (n = 6 treatment wells per time point). Relative to control (CTL)-treated cells, NAD3 treatments increased (p < 0.05) Sirt1 mRNA levels at 3 h, as well as global sirtuin activity at 3 and 24 h. Follow-up experiments comparing 24 h NAD3 or CTL treatments indicated that NAD3 increased nicotinamide phosphoribosyltransferase (NAMPT) and SIRT1 protein levels (p < 0.05). Cellular nicotinamide adenine dinucleotide (NAD+) levels were also elevated nearly two-fold after 24 h of NAD3 versus CTL treatments (p < 0.001). Markers of mitochondrial biogenesis were minimally affected. Although these data are limited to select biomarkers in vitro, these preliminary findings suggest that a theacrine-based supplement can modulate select biomarkers related to NAD+ biogenesis and sirtuin activity. However, these changes did not drive increases in mitochondrial biogenesis. While promising, these data are limited to a rodent cell line and human muscle biopsy studies are needed to validate and elucidate the significance of these findings.
Collapse
|
28
|
Gujarati NA, Vasquez JM, Bogenhagen DF, Mallipattu SK. The complicated role of mitochondria in the podocyte. Am J Physiol Renal Physiol 2020; 319:F955-F965. [PMID: 33073585 PMCID: PMC7792691 DOI: 10.1152/ajprenal.00393.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Mitochondria play a complex role in maintaining cellular function including ATP generation, generation of biosynthetic precursors for macromolecules, maintenance of redox homeostasis, and metabolic waste management. Although the contribution of mitochondrial function in various kidney diseases has been studied, there are still avenues that need to be explored under healthy and diseased conditions. Mitochondrial damage and dysfunction have been implicated in experimental models of podocytopathy as well as in humans with glomerular diseases resulting from podocyte dysfunction. Specifically, in the podocyte, metabolism is largely driven by oxidative phosphorylation or glycolysis depending on the metabolic needs. These metabolic needs may change drastically in the presence of podocyte injury in glomerular diseases such as diabetic kidney disease or focal segmental glomerulosclerosis. Here, we review the role of mitochondria in the podocyte and the factors regulating its function at baseline and in a variety of podocytopathies to identify potential targets for therapy.
Collapse
Affiliation(s)
- Nehaben A Gujarati
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Jessica M Vasquez
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Daniel F Bogenhagen
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
- Renal Section, Northport Department of Veterans Affairs Medical Center, Northport, New York
| |
Collapse
|
29
|
Zesiewicz TA, Hancock J, Ghanekar SD, Kuo SH, Dohse CA, Vega J. Emerging therapies in Friedreich's Ataxia. Expert Rev Neurother 2020; 20:1215-1228. [PMID: 32909841 PMCID: PMC8018609 DOI: 10.1080/14737175.2020.1821654] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Friedreich's ataxia (FRDA) is a progressive, neurodegenerative disease that results in gait and limb ataxia, diabetes, cardiac hypertrophy, and scoliosis. At the cellular level, FRDA results in the deficiency of frataxin, a mitochondrial protein that plays a vital role in iron homeostasis and amelioration of oxidative stress. No cure currently exists for FRDA, but exciting therapeutic developments which target different parts of the pathological cascade are on the horizon. AREAS COVERED Areas covered include past and emerging therapies for FRDA, including antioxidants and mitochondrial-related agents, nuclear factor erythroid-derived 2-related factor 2 (Nrf2) activators, deuterated polyunsaturated fatty acids, iron chelators, histone deacetylase (HDAC) inhibitors, trans-activator of transcription (TAT)-frataxin, interferon gamma (IFNγ), erythropoietin, resveratrol, gene therapy, and anti-sense oligonucleotides (ASOs), among others. EXPERT OPINION While drug discovery has been challenging, new and exciting prospective treatments for FRDA are currently on the horizon, including pharmaceutical agents and gene therapy. Agents that enhance mitochondrial function, such as Nrf2 activators, dPUFAs and catalytic antioxidants, as well as novel methods of frataxin augmentation and genetic modulation will hopefully provide treatment for this devastating disease.
Collapse
Affiliation(s)
- Theresa A. Zesiewicz
- University of South Florida (USF) Department of Neurology, USF Ataxia Research Center, Tampa Florida, James A Haley Veteran’s Hospital, Tampa, Florida, USA
| | - Joshua Hancock
- University of South Florida (USF) Department of Neurology, USF Ataxia Research Center, Tampa Florida, James A Haley Veteran’s Hospital, Tampa, Florida, USA
| | - Shaila D. Ghanekar
- University of South Florida (USF) Department of Neurology, USF Ataxia Research Center, Tampa Florida, James A Haley Veteran’s Hospital, Tampa, Florida, USA
| | - Sheng-Han Kuo
- Department of Neurology, Columbia University, New York, NY, USA
- Initiative for Columbia Ataxia and Tremor, New York, NY, USA
| | - Carlos A. Dohse
- Universidad Central Del Caribe School of Medicine, Bayamon, Puerto Rico
| | - Joshua Vega
- University of South Florida (USF) Department of Neurology, USF Ataxia Research Center, Tampa Florida, James A Haley Veteran’s Hospital, Tampa, Florida, USA
| |
Collapse
|
30
|
Human Mitochondrial Pathologies of the Respiratory Chain and ATP Synthase: Contributions from Studies of Saccharomyces cerevisiae. Life (Basel) 2020; 10:life10110304. [PMID: 33238568 PMCID: PMC7700678 DOI: 10.3390/life10110304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
The ease with which the unicellular yeast Saccharomyces cerevisiae can be manipulated genetically and biochemically has established this organism as a good model for the study of human mitochondrial diseases. The combined use of biochemical and molecular genetic tools has been instrumental in elucidating the functions of numerous yeast nuclear gene products with human homologs that affect a large number of metabolic and biological processes, including those housed in mitochondria. These include structural and catalytic subunits of enzymes and protein factors that impinge on the biogenesis of the respiratory chain. This article will review what is currently known about the genetics and clinical phenotypes of mitochondrial diseases of the respiratory chain and ATP synthase, with special emphasis on the contribution of information gained from pet mutants with mutations in nuclear genes that impair mitochondrial respiration. Our intent is to provide the yeast mitochondrial specialist with basic knowledge of human mitochondrial pathologies and the human specialist with information on how genes that directly and indirectly affect respiration were identified and characterized in yeast.
Collapse
|
31
|
Jeon YM, Kwon Y, Jo M, Lee S, Kim S, Kim HJ. The Role of Glial Mitochondria in α-Synuclein Toxicity. Front Cell Dev Biol 2020; 8:548283. [PMID: 33262983 PMCID: PMC7686475 DOI: 10.3389/fcell.2020.548283] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/20/2020] [Indexed: 12/18/2022] Open
Abstract
The abnormal accumulation of alpha-synuclein (α-syn) aggregates in neurons and glial cells is widely known to be associated with many neurodegenerative diseases, including Parkinson's disease (PD), Dementia with Lewy bodies (DLB), and Multiple system atrophy (MSA). Mitochondrial dysfunction in neurons and glia is known as a key feature of α-syn toxicity. Studies aimed at understanding α-syn-induced toxicity and its role in neurodegenerative diseases have primarily focused on neurons. However, a growing body of evidence demonstrates that glial cells such as microglia and astrocytes have been implicated in the initial pathogenesis and the progression of α-Synucleinopathy. Glial cells are important for supporting neuronal survival, synaptic functions, and local immunity. Furthermore, recent studies highlight the role of mitochondrial metabolism in the normal function of glial cells. In this work, we review the complex relationship between glial mitochondria and α-syn-mediated neurodegeneration, which may provide novel insights into the roles of glial cells in α-syn-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu-Mi Jeon
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Younghwi Kwon
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
- Department of Brain and Cognitive Sciences, DGIST, Daegu, South Korea
| | - Myungjin Jo
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Shinrye Lee
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Seyeon Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
- Department of Brain and Cognitive Sciences, DGIST, Daegu, South Korea
| | - Hyung-Jun Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| |
Collapse
|
32
|
Filanovsky K, Haran M, Mirkin V, Braester A, Shevetz O, Stanevsky A, Sigler E, Votinov E, Zaltsman-Amir Y, Berrebi A, Gross A, Shvidel L. Peripheral Blood Cell Mitochondrial Dysfunction in Myelodysplastic Syndrome Can Be Improved by a Combination of Coenzyme Q10 and Carnitine. Mediterr J Hematol Infect Dis 2020; 12:e2020072. [PMID: 33194146 PMCID: PMC7643803 DOI: 10.4084/mjhid.2020.072] [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: 06/20/2020] [Accepted: 10/03/2020] [Indexed: 11/08/2022] Open
Abstract
Structural mitochondrial abnormalities and genetic aberrations in mitochondrial proteins have been known in Myelodysplastic syndrome (MDS), yet there is currently little data regarding MDS's metabolic properties and energy production cells. In the current study, we used state-of-the-art methods to assess OXPHOS in peripheral blood cells obtained from MDS patients and healthy controls. We then assessed the effect of food supplements-Coenzyme Q10 and carnitine on mitochondrial function and hematological response. We show here for the first time that there is a significant impairment of mitochondrial respiration in peripheral blood cells in low-risk MDS, which can be improved with food supplements. We also show that these supplements may improve the cytopenia and quality of life.
Collapse
Affiliation(s)
- Kalman Filanovsky
- Hematology Institute, Kaplan medical center, Rehovot, Israel, affiliated with the Hebrew University, Jerusalem, Israel
| | - Michal Haran
- Hematology Institute, Kaplan medical center, Rehovot, Israel, affiliated with the Hebrew University, Jerusalem, Israel
| | - Vita Mirkin
- Hematology Institute, Kaplan medical center, Rehovot, Israel, affiliated with the Hebrew University, Jerusalem, Israel
| | - Andrei Braester
- Hematology institute, Galilee medical center, Nahariya, Israel
| | - Olga Shevetz
- Hematology Institute, Kaplan medical center, Rehovot, Israel, affiliated with the Hebrew University, Jerusalem, Israel
| | - Anfisa Stanevsky
- Hematology Institute, Kaplan medical center, Rehovot, Israel, affiliated with the Hebrew University, Jerusalem, Israel
| | - Erica Sigler
- Hematology Institute, Kaplan medical center, Rehovot, Israel, affiliated with the Hebrew University, Jerusalem, Israel
| | - Ekaterina Votinov
- Hematology Institute, Kaplan medical center, Rehovot, Israel, affiliated with the Hebrew University, Jerusalem, Israel
| | | | - Alain Berrebi
- Hematology Institute, Kaplan medical center, Rehovot, Israel, affiliated with the Hebrew University, Jerusalem, Israel
| | - Atan Gross
- Dept. of biological regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Lev Shvidel
- Hematology Institute, Kaplan medical center, Rehovot, Israel, affiliated with the Hebrew University, Jerusalem, Israel
| |
Collapse
|
33
|
Sun J, Patel CB, Jang T, Merchant M, Chen C, Kazerounian S, Diers AR, Kiebish MA, Vishnudas VK, Gesta S, Sarangarajan R, Narain NR, Nagpal S, Recht L. High levels of ubidecarenone (oxidized CoQ 10) delivered using a drug-lipid conjugate nanodispersion (BPM31510) differentially affect redox status and growth in malignant glioma versus non-tumor cells. Sci Rep 2020; 10:13899. [PMID: 32807842 PMCID: PMC7431533 DOI: 10.1038/s41598-020-70969-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 08/04/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolic reprogramming in cancer cells, vs. non-cancer cells, elevates levels of reactive oxygen species (ROS) leading to higher oxidative stress. The elevated ROS levels suggest a vulnerability to excess prooxidant loads leading to selective cell death, a therapeutically exploitable difference. Co-enzyme Q10 (CoQ10) an endogenous mitochondrial resident molecule, plays an important role in mitochondrial redox homeostasis, membrane integrity, and energy production. BPM31510 is a lipid-drug conjugate nanodispersion specifically formulated for delivery of supraphysiological concentrations of ubidecarenone (oxidized CoQ10) to the cell and mitochondria, in both in vitro and in vivo model systems. In this study, we sought to investigate the therapeutic potential of ubidecarenone in the highly treatment-refractory glioblastoma. Rodent (C6) and human (U251) glioma cell lines, and non-tumor human astrocytes (HA) and rodent NIH3T3 fibroblast cell lines were utilized for experiments. Tumor cell lines exhibited a marked increase in sensitivity to ubidecarenone vs. non-tumor cell lines. Further, elevated mitochondrial superoxide production was noted in tumor cells vs. non-tumor cells hours before any changes in proliferation or the cell cycle could be detected. In vitro co-culture experiments show ubidecarenone differentially affecting tumor cells vs. non-tumor cells, resulting in an equilibrated culture. In vivo activity in a highly aggressive orthotopic C6 glioma model demonstrated a greater than 25% long-term survival rate. Based on these findings we conclude that high levels of ubidecarenone delivered using BPM31510 provide an effective therapeutic modality targeting cancer-specific modulation of redox mechanisms for anti-cancer effects.
Collapse
Affiliation(s)
- Jiaxin Sun
- Department of Neurology and Clinical Neurosciences, Stanford University, Palo Alto, CA, 94305, USA.
| | - Chirag B Patel
- Department of Neurology and Clinical Neurosciences, Stanford University, Palo Alto, CA, 94305, USA.,Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Taichang Jang
- Department of Neurology and Clinical Neurosciences, Stanford University, Palo Alto, CA, 94305, USA
| | - Milton Merchant
- Department of Neurology and Clinical Neurosciences, Stanford University, Palo Alto, CA, 94305, USA
| | - Chen Chen
- Department of Otolaryngology, Stanford University, Palo Alto, CA, 94305, USA
| | | | | | | | | | | | | | | | - Seema Nagpal
- Department of Neurology and Clinical Neurosciences, Stanford University, Palo Alto, CA, 94305, USA
| | - Lawrence Recht
- Department of Neurology and Clinical Neurosciences, Stanford University, Palo Alto, CA, 94305, USA.
| |
Collapse
|
34
|
Cotta A, Alston CL, Baptista‐Junior S, Paim JF, Carvalho E, Navarro MM, Appleton M, Ng YS, Valicek J, da‐Cunha‐Junior AL, Lima MI, de la Rocque Ferreira A, Takata RI, Hargreaves IP, Gorman GS, McFarland R, Pierre G, Taylor RW. Early-onset coenzyme Q10 deficiency associated with ataxia and respiratory chain dysfunction due to novel pathogenic COQ8A variants, including a large intragenic deletion. JIMD Rep 2020; 54:45-53. [PMID: 32685350 PMCID: PMC7358671 DOI: 10.1002/jmd2.12107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/25/2020] [Accepted: 02/11/2020] [Indexed: 11/13/2022] Open
Abstract
Coenzyme Q10 (CoQ10) deficiency is a clinically and genetically heterogeneous subtype of mitochondrial disease. We report two girls with ataxia and mitochondrial respiratory chain deficiency who were shown to have primary CoQ10 deficiency. Muscle histochemistry displayed signs of mitochondrial dysfunction-ragged red fibers, mitochondrial paracrystalline inclusions, and lipid deposits while biochemical analyses revealed complex II+III respiratory chain deficiencies. MRI brain demonstrated cerebral and cerebellar atrophy. Targeted molecular analysis identified a homozygous c.1015G>A, p.(Ala339Thr) COQ8A variant in subject 1, while subject 2 was found to harbor a single heterozygous c.1029_1030delinsCA variant predicting a p.Gln343_Val344delinsHisMet amino acid substitution. Subsequent investigations identified a large-scale COQ8A deletion in trans to the c.1029_1030delinsCA allele. A skin biopsy facilitated cDNA studies that confirmed exon skipping in the fibroblast derived COQ8A mRNA transcript. This report expands the molecular genetic spectrum associated with COQ8A-related mitochondrial disease and highlights the importance of thorough investigation of candidate pathogenic variants to establish phase. Rapid diagnosis is of the utmost importance as patients may benefit from therapeutic CoQ10 supplementation.
Collapse
Affiliation(s)
- Ana Cotta
- Department of PathologySARAH Network of Rehabilitation HospitalsBelo HorizonteBrazil
| | - Charlotte L. Alston
- Wellcome Centre for Mitochondrial ResearchTranslational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle UniversityNewcastle upon TyneUK
- NHS Highly Specialised Services for Rare Mitochondrial Disorders, Royal Victoria InfirmaryNewcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | | | - Julia F. Paim
- Department of PathologySARAH Network of Rehabilitation HospitalsBelo HorizonteBrazil
| | - Elmano Carvalho
- Department of NeurophysiologySARAH Network of Rehabilitation HospitalsBelo HorizonteBrazil
| | - Monica M. Navarro
- Department of Pediatrics and GeneticsSARAH Network of Rehabilitation HospitalsBelo HorizonteBrazil
| | - Marie Appleton
- Clinical Biochemistry, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Yi Shiau Ng
- Wellcome Centre for Mitochondrial ResearchTranslational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle UniversityNewcastle upon TyneUK
- NHS Highly Specialised Services for Rare Mitochondrial Disorders, Royal Victoria InfirmaryNewcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Jaquelin Valicek
- Department of NeurophysiologySARAH Network of Rehabilitation HospitalsBelo HorizonteBrazil
| | | | - Maria I. Lima
- Department of Electron MicroscopySARAH Network of Rehabilitation HospitalsBrasíliaBrazil
| | | | - Reinaldo I. Takata
- Department of Molecular BiologySARAH Network of Rehabilitation HospitalsBrasíliaBrazil
| | - Iain P. Hargreaves
- Neurometabolic UnitNational Hospital for Neurology and NeurosurgeryLondonUK
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores UniversityLiverpoolUK
| | - Gráinne S. Gorman
- Wellcome Centre for Mitochondrial ResearchTranslational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle UniversityNewcastle upon TyneUK
- NHS Highly Specialised Services for Rare Mitochondrial Disorders, Royal Victoria InfirmaryNewcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Robert McFarland
- Wellcome Centre for Mitochondrial ResearchTranslational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle UniversityNewcastle upon TyneUK
- NHS Highly Specialised Services for Rare Mitochondrial Disorders, Royal Victoria InfirmaryNewcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Germaine Pierre
- South West Regional Metabolic DepartmentBristol Royal Hospital for ChildrenBristolUK
| | - Robert W. Taylor
- Wellcome Centre for Mitochondrial ResearchTranslational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle UniversityNewcastle upon TyneUK
- NHS Highly Specialised Services for Rare Mitochondrial Disorders, Royal Victoria InfirmaryNewcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| |
Collapse
|
35
|
Justine Perrin R, Rousset‐Rouvière C, Garaix F, Cano A, Conrath J, Boyer O, Tsimaratos M. COQ6 mutation in patients with nephrotic syndrome, sensorineural deafness, and optic atrophy. JIMD Rep 2020; 54:37-44. [PMID: 32685349 PMCID: PMC7358665 DOI: 10.1002/jmd2.12068] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/18/2019] [Accepted: 06/24/2019] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Primary coenzyme Q10 (CoQ10) deficiencies are a group of mitochondrial disorders that has proven responsiveness to replacement therapy. Mutations in enzymes involved in the biosynthesis of CoQ10 genes are associated with these deficits. The clinical presentation of this rare autosomal recessive disorder is heterogeneous and depends on the gene involved. Mutations in the COQ2, COQ6, PDSS2, and ADCK4 genes are responsible for steroid-resistant nephrotic syndrome (SRNS), which is associated with extra-renal symptoms. Previous studies have reported COQ6 mutations in 11 patients from five different families presenting with SRNS and sensorineural deafness. CASE REPORTS Our study reports the cases of two brothers of Turkish origin with renal failure and sensorineural deafness associated with COQ6 mutations responsible of CoQ10 deficiency. Optical symptoms were present in the eldest, that improved with Idebenone. CONCLUSION/DISCUSSION For the first time, COQ6 mutation with optical involvement is associated with renal and hearing impairment. Although the response to replacement CoQ10 therapy was difficult to evaluate, we think that this treatment was able to stop the disease progression in both patients, and even to prevent the occurrence/development of optical and neurological impairment in the younger brother. Mitochondrial dysfunction secondary to CoQ10 deficiency should always be suspected in patients with SRNS and extra-renal symptoms. Early recognition of this genetic SRNS is mandatory since SRNS can be avoided by adequate treatment based on CoQ10 supplement or an analogue. All cases of primary CoQ10 deficiency should be treated at an early stage to limit the progression of lesions and prevent the emergence of new symptoms.
Collapse
Affiliation(s)
- R. Justine Perrin
- Assistance Public Hôpitaux de MarseilleService Multidisciplinaire TimoneMarseilleFrance
| | | | - Florentine Garaix
- Assistance Public Hôpitaux de MarseilleService Multidisciplinaire TimoneMarseilleFrance
| | - Aline Cano
- Assistance Public Hôpitaux de MarseilleService de pédiatrie et NeurologieMarseilleFrance
| | - John Conrath
- Clinique Monticelli, OphtalmologieMarseilleFrance
| | - Olivia Boyer
- Hopital NeckerNéphrologie PediatriqueParisFrance
| | - Michel Tsimaratos
- Assistance Public Hôpitaux de MarseilleService Multidisciplinaire TimoneMarseilleFrance
| |
Collapse
|
36
|
Gana S, Valente EM. Movement Disorders in Genetic Pediatric Ataxias. Mov Disord Clin Pract 2020; 7:383-393. [PMID: 32373654 DOI: 10.1002/mdc3.12937] [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: 11/19/2019] [Revised: 02/24/2020] [Accepted: 03/08/2020] [Indexed: 11/06/2022] Open
Abstract
Background Genetic pediatric ataxias are heterogeneous rare disorders, mainly inherited as autosomal-recessive traits. Most forms are progressive and lack effective treatment, with relevant socioeconomical impact. Albeit ataxia represents the main clinical feature, the phenotype can be more complex, with additional neurological and nonneurological signs being described in several forms. Methods and Results In this review, we provide an overview of the occurrence and spectrum of movement disorders in the most relevant forms of childhood-onset genetic ataxias. All types of hypokinetic and hyperkinetic movement disorders of variable severity have been reported. Movement disorders occasionally represent the symptom of onset, predating ataxia even of a few years and therefore challenging an early diagnosis. Their pathogenesis still remains poorly defined, as it is not yet clear whether movement disorders may directly relate to the cerebellar pathology or result from an extracerebellar dysfunction, including the basal ganglia. Conclusion Recognition of the complete movement disorder phenotype in genetic pediatric ataxias has important implications for diagnosis, management, and genetic counseling.
Collapse
Affiliation(s)
| | - Enza Maria Valente
- IRCCS Mondino Foundation Pavia Italy.,Department of Molecular Medicine University of Pavia Pavia Italy
| |
Collapse
|
37
|
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
|
38
|
Berenguel Hernández AM, de la Cruz M, Alcázar-Fabra M, Prieto-Rodríguez A, Sánchez-Cuesta A, Martin J, Tormo JR, Rodríguez-Aguilera JC, Cortés-Rodríguez AB, Navas P, Reyes F, Vicente F, Genilloud O, Santos-Ocaña C. Design of High-Throughput Screening of Natural Extracts to Identify Molecules Bypassing Primary Coenzyme Q Deficiency in Saccharomyces cerevisiae. SLAS DISCOVERY 2019; 25:299-309. [PMID: 31751168 DOI: 10.1177/2472555219877185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Coenzyme Q10 (CoQ10) deficiency syndrome is a rare disease included in the family of mitochondrial diseases, which is a heterogeneous group of genetic disorders characterized by defective energy production. CoQ10 biosynthesis in humans requires at least 11 gene products acting in a multiprotein complex within mitochondria. The high-throughput screening (HTS) method based on the stabilization of the CoQ biosynthesis complex (Q-synthome) produced by the COQ8 gene overexpression is proven here to be a successful method for identifying new molecules from natural extracts that are able to bypass the CoQ6 deficiency in yeast mutant cells. The main features of the new approach are the combination of two yeast targets defective in genes with different functions on CoQ6 biosynthesis to secure the versatility of the molecule identified, the use of glycerol as a nonfermentable carbon source providing a wide growth window, and the stringent conditions required to mark an extract as positive. The application of this pilot approach to a representative subset of 1200 samples of the Library of Natural Products of Fundación MEDINA resulted in the finding of nine positive extracts. The fractionation of three of the nine extracts allowed the identification of five molecules; two of them are present in molecule databases of natural extracts and three are nondescribed molecules. The use of this screening method opens the possibility of discovering molecules with CoQ10-bypassing action useful as therapeutic agents to fight against mitochondrial diseases in human patients.
Collapse
Affiliation(s)
| | | | - María Alcázar-Fabra
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Sevilla, Spain.,CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Ana Sánchez-Cuesta
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Sevilla, Spain.,CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Juan Carlos Rodríguez-Aguilera
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Sevilla, Spain.,CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Belén Cortés-Rodríguez
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Sevilla, Spain.,CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Plácido Navas
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Sevilla, Spain.,CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | | | - Carlos Santos-Ocaña
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Sevilla, Spain.,CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
39
|
Galosi S, Barca E, Carrozzo R, Schirinzi T, Quinzii CM, Lieto M, Vasco G, Zanni G, Di Nottia M, Galatolo D, Filla A, Bertini E, Santorelli FM, Leuzzi V, Haas R, Hirano M, Friedman J. Dystonia-Ataxia with early handwriting deterioration in COQ8A mutation carriers: A case series and literature review. Parkinsonism Relat Disord 2019; 68:8-16. [DOI: 10.1016/j.parkreldis.2019.09.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/27/2019] [Accepted: 09/15/2019] [Indexed: 02/06/2023]
|
40
|
Bianco M, Lopes JA, Beiral HJV, Filho JDD, Frankenfeld SP, Fortunato RS, Gattass CR, Vieyra A, Takiya CM. The contralateral kidney presents with impaired mitochondrial functions and disrupted redox homeostasis after 14 days of unilateral ureteral obstruction in mice. PLoS One 2019; 14:e0218986. [PMID: 31251767 PMCID: PMC6599136 DOI: 10.1371/journal.pone.0218986] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 06/14/2019] [Indexed: 02/07/2023] Open
Abstract
In unilateral ureteral obstruction (UUO), both oxidative stress and mitochondrial dysfunction are related to cell death. The aim of this study has been to characterize profiles of enzyme antioxidant activities and mitochondrial functioning of the contralateral (CL) compared to UUO and Sham (false-operated) kidneys of Balb/c mice. Kidneys were resected 14 days after obstruction for immunohistochemical and cortical mitochondrial functioning assays. Antioxidant enzymes activities were investigated in mitochondria and cytosol. Oxygen consumption (QO2) and formation of O2 reactive species (ROS) were assessed with pyruvate plus malate or succinate as the respiratory substrates. QO2 decreased in CL and UUO in all states using substrates for complex II, whereas it was affected only in UUO when substrates for complex I were used. Progressive decrease in mitochondrial ROS formation–in the forward and reverse pathway at complex I–correlates well with the inhibition of QO2 and, therefore, with decreased electron transfer at the level of complexes upstream of cytochrome c oxidase. CL and UUO transmembrane potential responses to ADP were impaired with succinate. Intense Ca2+-induced swelling was elicited in CL and UUO mitochondria. Important and selective differences exist in CL antioxidant enzymes with respect to either Sham or UUO kidneys: CL kidneys had increased mitochondrial glutathione peroxidase and cytosolic catalase activities, indicative of compensatory responses in the face of an early altered ROS homeostasis (as detected by 4-hydroxynonenal), and of a significant tendency to apoptosis. In CL and UUO, upregulation of nuclear (erythroid-derived 2)-like 2 transcription factor (Nrf2), as well as of cytoplasmic and nuclear Kelch-like ECH-associated protein 1 (Keap1) in opposition to decreased heme oxygenase-1 (HO-1), suggest impairment of the Nrf2/Keap1/HO-1 system. It is concluded that chronic obstruction impairs mitochondrial function in CL and UUO, preferentially affecting complex II.
Collapse
Affiliation(s)
- Mario Bianco
- Center of Experimental Surgery, Postgraduate Program in Surgical Science, Department of Surgery, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| | - Jarlene A. Lopes
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hellen J. V. Beiral
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - João D. D. Filho
- Center of Experimental Surgery, Postgraduate Program in Surgical Science, Department of Surgery, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Stephan P. Frankenfeld
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo S. Fortunato
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cerli R. Gattass
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging /CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Translational Biomedicine Program, Grande Rio University, Duque de Caxias, Brazil
| | - Christina M. Takiya
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
41
|
Miniaturized imprinted solid phase extraction to the selective analysis of Coenzyme Q10 in urine. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1116:24-29. [DOI: 10.1016/j.jchromb.2019.03.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 11/24/2022]
|
42
|
Cerqua C, Casarin A, Pierrel F, Vazquez Fonseca L, Viola G, Salviati L, Trevisson E. Vitamin K2 cannot substitute Coenzyme Q 10 as electron carrier in the mitochondrial respiratory chain of mammalian cells. Sci Rep 2019; 9:6553. [PMID: 31024065 PMCID: PMC6484000 DOI: 10.1038/s41598-019-43014-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/11/2019] [Indexed: 12/16/2022] Open
Abstract
Coenzyme Q10 (CoQ10) deficiencies are a group of heterogeneous conditions that respond to ubiquinone administration if treated soon after the onset of symptoms. However, this treatment is only partially effective due to its poor bioavailability. We tested whether vitamin K2, which was reported to act as a mitochondrial electron carrier in D. melanogaster, could mimic ubiquinone function in human CoQ10 deficient cell lines, and in yeast carrying mutations in genes required for coenzyme Q6 (CoQ6) biosynthesis. We found that vitamin K2, despite entering into mitochondria, restored neither electron flow in the respiratory chain, nor ATP synthesis. Conversely, coenzyme Q4 (CoQ4), an analog of CoQ10 with a shorter isoprenoid side chain, could efficiently substitute its function. Given its better solubility, CoQ4 could represent an alternative to CoQ10 in patients with both primary and secondary CoQ10 deficiencies.
Collapse
Affiliation(s)
- Cristina Cerqua
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy.,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy
| | - Alberto Casarin
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy.,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy
| | - Fabien Pierrel
- Univ. Grenoble Alpes, CNRS, CHU Grenoble Alpes, Grenoble INP, TIMC-IMAG, 38000, Grenoble, France
| | - Luis Vazquez Fonseca
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy.,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy
| | - Giampiero Viola
- Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.,Pediatric Hematooncology Laboratory, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy
| | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy. .,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.
| | - Eva Trevisson
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy. .,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.
| |
Collapse
|
43
|
Fracassi A, Marangoni M, Rosso P, Pallottini V, Fioramonti M, Siteni S, Segatto M. Statins and the Brain: More than Lipid Lowering Agents? Curr Neuropharmacol 2019; 17:59-83. [PMID: 28676012 PMCID: PMC6341496 DOI: 10.2174/1570159x15666170703101816] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/24/2017] [Accepted: 06/26/2017] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Statins represent a class of medications widely prescribed to efficiently treat dyslipidemia. These drugs inhibit 3-βhydroxy 3β-methylglutaryl Coenzyme A reductase (HMGR), the rate-limiting enzyme of mevalonate (MVA) pathway. Besides cholesterol, MVA pathway leads to the production of several other compounds, which are essential in the regulation of a plethora of biological activities, including in the central nervous system. For these reasons, statins are able to induce pleiotropic actions, and acquire increased interest as potential and novel modulators in brain processes, especially during pathological conditions. OBJECTIVE The purpose of this review is to summarize and examine the current knowledge about pharmacokinetic and pharmacodynamic properties of statins in the brain. In addition, effects of statin on brain diseases are discussed providing the most up-to-date information. METHODS Relevant scientific information was identified from PubMed database using the following keywords: statins and brain, central nervous system, neurological diseases, neurodegeneration, brain tumors, mood, stroke. RESULTS 315 scientific articles were selected and analyzed for the writing of this review article. Several papers highlighted that statin treatment is effective in preventing or ameliorating the symptomatology of a number of brain pathologies. However, other studies failed to demonstrate a neuroprotective effect. CONCLUSION Even though considerable research studies suggest pivotal functional outcomes induced by statin therapy, additional investigation is required to better determine the pharmacological effectiveness of statins in the brain, and support their clinical use in the management of different neuropathologies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marco Segatto
- Address correspondence to this author at the Department of Sense Organs, Sapienza University, viale del Policlinico 155, 00186 Rome, Italy; E-mail:
| |
Collapse
|
44
|
Chen FL, Chang PS, Lin YC, Lin PT. A Pilot Clinical Study of Liquid Ubiquinol Supplementation on Cardiac Function in Pediatric Dilated Cardiomyopathy. Nutrients 2018; 10:nu10111697. [PMID: 30405022 PMCID: PMC6266971 DOI: 10.3390/nu10111697] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/25/2018] [Accepted: 11/05/2018] [Indexed: 11/24/2022] Open
Abstract
Background: Pediatric dilated cardiomyopathy (PDCM) is a life-threatening type of cardiac muscle dysfunction in children. Ubiquinone is a lipid-soluble nutrient that participates in energy synthesis. Recently, a novel hydrophilic ubiquinol supplement was developed. The purpose of this study was to assess the effect of liquid ubiquinol supplementation (10 mg/kg body weight/day) on cardiac function in children with PDCM. Methods: Ten children diagnosed with PDCM were recruited to this study and administered with liquid ubiquinol for 24 weeks. The cardiac function was measured by echocardiography. The New York Heart Association (NYHA) functional classification was used to assess symptoms of heart failure. Plasma coenzyme Q10 levels were measured during the study. Results: Ejection fraction (EF) and fractional shortening (FS) were significantly higher than the baseline values until week 16 of supplementation. Subjects who had higher plasma coenzyme Q10 concentration had significantly better EF and FS values. In addition, 30% of the subjects showed improvement in the NYHA classification after 24 weeks of supplementation. Conclusion: Liquid ubiquinol supplementation is associated with an increase the level of coenzyme Q10 to complementary improve cardiac function (particularly EF and FS) and ameliorate the symptoms of heart failure in children with PDCM.
Collapse
Affiliation(s)
- Fong-Lin Chen
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
- Pediatric Cardiology Clinic, Taichung 40244, Taiwan.
| | - Po-Sheng Chang
- Department of Nutrition, Chung Shan Medical University, Taichung 40201, Taiwan.
- Graduate Program in Nutrition, Chung Shan Medical University, Taichung 40201, Taiwan.
| | - Yi-Chin Lin
- Department of Nutrition, Chung Shan Medical University, Taichung 40201, Taiwan.
| | - Ping-Ting Lin
- Department of Nutrition, Chung Shan Medical University, Taichung 40201, Taiwan.
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
| |
Collapse
|
45
|
Pandey R, Riley CL, Mills EM, Tiziani S. Highly sensitive and selective determination of redox states of coenzymes Q 9 and Q 10 in mice tissues: Application of orbitrap mass spectrometry. Anal Chim Acta 2018; 1011:68-76. [PMID: 29475487 DOI: 10.1016/j.aca.2018.01.066] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 12/31/2022]
Abstract
Coenzyme Q (CoQ) is a redox active molecule that plays a fundamental role in mitochondrial energy generation and functions as a potent endogenous antioxidant. Redox ratio of CoQ has been suggested as a good marker of mitochondrial dysfunction and oxidative stress. Nevertheless, simultaneous measurement of redox states of CoQ is challenging owing to its hydrophobicity and instability of the reduced form. In order to improve the analytical methodology, paying special attention to this instability, we developed a highly sensitive and selective high-resolution/accurate-mass (HR/AM) UHPLC-MS/MS method for the rapid determination of redox states of CoQ9 and CoQ10 by ultra-performance liquid chromatography-hybrid quadrupole-Orbitrap mass spectrometry. CoQs were extracted using hexane with the addition of butylated hydroxytoluene to limit oxidation during sample preparation. Chromatographic separation of the analytes was achieved on a Kinetex C18 column with the isocratic elution of 5 mM ammonium formate in 2-propanol/methanol (60:40) within 4 min. A full MS/all ion fragmentation (AIF) acquisition mode with mass accuracy < 5 ppm was used for detection and determination of redox states of CoQ9 and CoQ10 in healthy mice tissues using reduced and oxidized CoQ4 as internal standards. The validated method showed good linearity (r2 ≥ 0.9991), intraday, inter-day precision (CVs ≤ 11.9%) and accuracy (RE ≤±15.2%). In contrast to existing methods, the current method offers enhanced sensitivity (up to 52 fold) with LOD and LOQ ranged from 0.01 to 0.49 ng mL-1 and 0.04-1.48 ng mL-1, respectively. Moreover, we evaluated various diluents to investigate bench top stability (at 4 °C) of targeted analytes in tissue samples during LC-MS assay up to 24 h. Ethanol was determined to be an optimum diluent without any significant oxidation of reduced CoQ up to 24 h. The developed method offers a rapid, highly sensitive and selective strategy for the measurement of redox states of CoQs in clinical studies.
Collapse
Affiliation(s)
- Renu Pandey
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX 78712, USA; Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Christopher L Riley
- Department of Molecular Biosciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Edward M Mills
- Division of Pharmacy and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Stefano Tiziani
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX 78712, USA; Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
46
|
Parikh S, Goldstein A, Karaa A, Koenig MK, Anselm I, Brunel-Guitton C, Christodoulou J, Cohen BH, Dimmock D, Enns GM, Falk MJ, Feigenbaum A, Frye RE, Ganesh J, Griesemer D, Haas R, Horvath R, Korson M, Kruer MC, Mancuso M, McCormack S, Raboisson MJ, Reimschisel T, Salvarinova R, Saneto RP, Scaglia F, Shoffner J, Stacpoole PW, Sue CM, Tarnopolsky M, Van Karnebeek C, Wolfe LA, Cunningham ZZ, Rahman S, Chinnery PF. Patient care standards for primary mitochondrial disease: a consensus statement from the Mitochondrial Medicine Society. Genet Med 2017; 19:S1098-3600(21)04766-3. [PMID: 28749475 PMCID: PMC7804217 DOI: 10.1038/gim.2017.107] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/25/2017] [Indexed: 02/07/2023] Open
Abstract
The purpose of this statement is to provide consensus-based recommendations for optimal management and care for patients with primary mitochondrial disease. This statement is intended for physicians who are engaged in the diagnosis and management of these patients. Working group members were appointed by the Mitochondrial Medicine Society. The panel included members with several different areas of expertise. The panel members utilized surveys and the Delphi method to reach consensus. We anticipate that this statement will need to be updated as the field continues to evolve. Consensus-based recommendations are provided for the routine care and management of patients with primary genetic mitochondrial disease.
Collapse
Affiliation(s)
- Sumit Parikh
- Center for Child Neurology, Cleveland Clinic Children’s Hospital, Cleveland, Ohio, USA
| | - Amy Goldstein
- Division of Child Neurology, Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amel Karaa
- Division of Genetics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mary Kay Koenig
- Division of Child and Adolescent Neurology, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Irina Anselm
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts, USA
| | | | - John Christodoulou
- Neurodevelopmental Genomics Research Group, Murdoch Childrens Research Institute, and Department of Paediatrics, Melbourne Medical School, University of Melbourne, Melbourne, Australia
| | - Bruce H. Cohen
- Neurodevelopmental Science Center, Children’s Hospital Medical Center of Akron, Akron, Ohio, USA
| | - David Dimmock
- Rady Children’s Institute for Genomic Medicine, San Diego, California, USA
| | - Gregory M. Enns
- Division of Medical Genetics, Department of Pediatrics, Stanford University Lucile Packard Children’s Hospital, Palo Alto, California, USA
| | - Marni J. Falk
- Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Annette Feigenbaum
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, University of California San Diego and Rady Childrens Hospital, San Diego, California, USA
| | - Richard E. Frye
- Department of Pediatrics, University of Arkansas Medical Sciences, Little Rock, Arkansas, USA
| | - Jaya Ganesh
- Division of Genetics, Department of Pediatrics, Cooper Medical School at Rowan University, Camden, New Jersey, USA
| | - David Griesemer
- Division of Neurology, Levine Children’s Hospital, Charlotte, North Carolina, USA
| | - Richard Haas
- Departments of Neurosciences and Pediatrics, University of California San Diego, La Jolla, California, USA
- Department of Neurosciences, Rady Children’s Hospital, San Diego, California, USA
| | - Rita Horvath
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Mark Korson
- Genetic Metabolic Center for Education, Salem, Massachusetts, USA
| | - Michael C. Kruer
- Department of Pediatric Neurology, University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Michelangelo Mancuso
- Department of Experimental and Clinical Medicine, Neurological Clinic, University of Pisa, Pisa, Italy
| | - Shana McCormack
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Tyler Reimschisel
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ramona Salvarinova
- Division of Biochemical Diseases, BC Children’s Hospital, British Columbia, Canada
| | - Russell P. Saneto
- Department of Neurology, Seattle Children’s Hospital/University of Washington, Seattle, Washington, USA
| | - Fernando Scaglia
- Department of Molecular and Human Genetics, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas, USA
| | - John Shoffner
- Neurology, Biochemical & Molecular Genetics, Atlanta, Georgia, USA
| | - Peter W. Stacpoole
- Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Carolyn M. Sue
- Department of Neurology and Kolling Institute, Royal North Shore Hospital, St Leonards, Australia
| | - Mark Tarnopolsky
- Division of Neurology, McMaster University, Hamilton, Ontario, Canada
| | - Clara Van Karnebeek
- Department of Pediatrics, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatrics, Centre for Molecular Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lynne A. Wolfe
- Undiagnosed Diseases Network, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Shamima Rahman
- Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Patrick F. Chinnery
- Department of Clinical Neurosciences & MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Nochi Z, Olsen RKJ, Gregersen N. Short-chain acyl-CoA dehydrogenase deficiency: from gene to cell pathology and possible disease mechanisms. J Inherit Metab Dis 2017; 40:641-655. [PMID: 28516284 DOI: 10.1007/s10545-017-0047-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 03/31/2017] [Accepted: 04/05/2017] [Indexed: 12/15/2022]
Abstract
Short-chain acyl-CoA dehydrogenase deficiency (SCADD) is an inherited disorder of mitochondrial fatty acid oxidation that is characterized by the presence of increased butyrylcarnitine and ethylmalonic acid (EMA) concentrations in plasma and urine. Individuals with symptomatic SCADD may show relatively severe phenotype, while the majority of those who are diagnosed through newborn screening by tandem mass spectrometry may remain asymptomatic. As such, the associated clinical symptoms are very diverse, ranging from severe metabolic or neuromuscular disabilities to asymptomatic. Molecular analysis of affected individuals has identified rare gene variants along with two common gene variants, c.511C > T and c.625G > A. In vitro studies have demonstrated that the common variants as well as the great majority of rare variants, which are missense variants, impair folding, that may lead to toxic accumulation of the encoded protein, and/or metabolites, and initiate excessive production of ROS and chronic oxidative stress. It has been suggested that this cell toxicity in combination with yet unknown factors can trigger disease development. This association and the full implications of SCADD are not commonly appreciated. Accordingly, there is a worldwide discussion of the relationship of clinical manifestation to SCADD, and whether SCAD gene variants are disease associated at all. Therefore, SCADD is not part of the newborn screening programs in most countries, and consequently many patients with SCAD gene variants do not get a diagnosis and the possibilities to be followed up during development.
Collapse
Affiliation(s)
- Zahra Nochi
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University Hospital and Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark.
| | - Rikke Katrine Jentoft Olsen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University Hospital and Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | - Niels Gregersen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University Hospital and Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| |
Collapse
|
48
|
Romero-Moya D, Santos-Ocaña C, Castaño J, Garrabou G, Rodríguez-Gómez JA, Ruiz-Bonilla V, Bueno C, González-Rodríguez P, Giorgetti A, Perdiguero E, Prieto C, Moren-Nuñez C, Fernández-Ayala DJ, Victoria Cascajo M, Velasco I, Canals JM, Montero R, Yubero D, Jou C, López-Barneo J, Cardellach F, Muñoz-Cánoves P, Artuch R, Navas P, Menendez P. Genetic Rescue of Mitochondrial and Skeletal Muscle Impairment in an Induced Pluripotent Stem Cells Model of Coenzyme Q 10 Deficiency. Stem Cells 2017; 35:1687-1703. [PMID: 28472853 DOI: 10.1002/stem.2634] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/29/2017] [Accepted: 04/12/2017] [Indexed: 02/06/2023]
Abstract
Coenzyme Q10 (CoQ10 ) plays a crucial role in mitochondria as an electron carrier within the mitochondrial respiratory chain (MRC) and is an essential antioxidant. Mutations in genes responsible for CoQ10 biosynthesis (COQ genes) cause primary CoQ10 deficiency, a rare and heterogeneous mitochondrial disorder with no clear genotype-phenotype association, mainly affecting tissues with high-energy demand including brain and skeletal muscle (SkM). Here, we report a four-year-old girl diagnosed with minor mental retardation and lethal rhabdomyolysis harboring a heterozygous mutation (c.483G > C (E161D)) in COQ4. The patient's fibroblasts showed a decrease in [CoQ10 ], CoQ10 biosynthesis, MRC activity affecting complexes I/II + III, and respiration defects. Bona fide induced pluripotent stem cell (iPSCs) lines carrying the COQ4 mutation (CQ4-iPSCs) were generated, characterized and genetically edited using the CRISPR-Cas9 system (CQ4ed -iPSCs). Extensive differentiation and metabolic assays of control-iPSCs, CQ4-iPSCs and CQ4ed -iPSCs demonstrated a genotype association, reproducing the disease phenotype. The COQ4 mutation in iPSC was associated with CoQ10 deficiency, metabolic dysfunction, and respiration defects. iPSC differentiation into SkM was compromised, and the resulting SkM also displayed respiration defects. Remarkably, iPSC differentiation in dopaminergic or motor neurons was unaffected. This study offers an unprecedented iPSC model recapitulating CoQ10 deficiency-associated functional and metabolic phenotypes caused by COQ4 mutation. Stem Cells 2017;35:1687-1703.
Collapse
Affiliation(s)
- Damià Romero-Moya
- Josep Carreras Leukemia Research Institute, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Carlos Santos-Ocaña
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo Olavide-CSIC, Sevilla, Spain.,CIBER de Enfermedades Raras (CIBERER), Spain
| | - Julio Castaño
- Josep Carreras Leukemia Research Institute, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Gloria Garrabou
- CIBER de Enfermedades Raras (CIBERER), Spain.,Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS-Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - José A Rodríguez-Gómez
- Institute of Biomedicine of Seville, Hospital Universitario Virgen del Rocío-Consejo Superior de Investigaciones Científicas (CSIC)-University of Seville, Seville, Spain
| | - Vanesa Ruiz-Bonilla
- CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Pompeu Fabra University (UPF), Barcelona, Spain
| | - Clara Bueno
- Josep Carreras Leukemia Research Institute, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Patricia González-Rodríguez
- Institute of Biomedicine of Seville, Hospital Universitario Virgen del Rocío-Consejo Superior de Investigaciones Científicas (CSIC)-University of Seville, Seville, Spain.,CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Alessandra Giorgetti
- Josep Carreras Leukemia Research Institute, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Eusebio Perdiguero
- CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Pompeu Fabra University (UPF), Barcelona, Spain
| | - Cristina Prieto
- Josep Carreras Leukemia Research Institute, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Constanza Moren-Nuñez
- CIBER de Enfermedades Raras (CIBERER), Spain.,Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS-Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Daniel J Fernández-Ayala
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo Olavide-CSIC, Sevilla, Spain.,CIBER de Enfermedades Raras (CIBERER), Spain
| | - Maria Victoria Cascajo
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo Olavide-CSIC, Sevilla, Spain.,CIBER de Enfermedades Raras (CIBERER), Spain
| | - Iván Velasco
- Insituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México.,Laboratorio de Reprogramación Celular del IFC en el Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", México DF, México
| | - Josep Maria Canals
- CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Stem Cells and Regenerative Medicine Laboratory, Production and validation center of advanced therapies (Creatio) Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Neuroscience Institute, University of Barcelona, Barcelona, Spain.,August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Raquel Montero
- CIBER de Enfermedades Raras (CIBERER), Spain.,Clinical Biochemistry Department, Pediatric Research Institute-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Delia Yubero
- Clinical Biochemistry Department, Pediatric Research Institute-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Cristina Jou
- CIBER de Enfermedades Raras (CIBERER), Spain.,Clinical Biochemistry Department, Pediatric Research Institute-Hospital Sant Joan de Déu, Barcelona, Spain
| | - José López-Barneo
- Institute of Biomedicine of Seville, Hospital Universitario Virgen del Rocío-Consejo Superior de Investigaciones Científicas (CSIC)-University of Seville, Seville, Spain.,CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Francesc Cardellach
- CIBER de Enfermedades Raras (CIBERER), Spain.,Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS-Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Pura Muñoz-Cánoves
- CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Pompeu Fabra University (UPF), Barcelona, Spain.,Institució Catalana Recerca Estudis Avančats (ICREA), Lluís Companys 23, Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| | - Rafael Artuch
- CIBER de Enfermedades Raras (CIBERER), Spain.,Clinical Biochemistry Department, Pediatric Research Institute-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Plácido Navas
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo Olavide-CSIC, Sevilla, Spain.,CIBER de Enfermedades Raras (CIBERER), Spain
| | - Pablo Menendez
- Josep Carreras Leukemia Research Institute, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Institució Catalana Recerca Estudis Avančats (ICREA), Lluís Companys 23, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCIII, Spain
| |
Collapse
|
49
|
Coenzyme Q10 attenuated β-amyloid 25–35 –induced inflammatory responses in PC12 cells through regulation of the NF–κB signaling pathway. Brain Res Bull 2017; 131:192-198. [DOI: 10.1016/j.brainresbull.2017.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/24/2017] [Indexed: 11/30/2022]
|
50
|
Wang HM, Yang HL, Thiyagarajan V, Huang TH, Huang PJ, Chen SC, Liu JY, Hsu LS, Chang HW, Hseu YC. Coenzyme Q 0 Enhances Ultraviolet B-Induced Apoptosis in Human Estrogen Receptor-Positive Breast (MCF-7) Cancer Cells. Integr Cancer Ther 2016; 16:385-396. [PMID: 27821721 PMCID: PMC5759941 DOI: 10.1177/1534735416673907] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Coenzyme Q0 (CoQ0; 2,3-dimethoxy-5-methyl-1,4-benzoquinone), a major active constituent of Antrodia camphorata, has been shown to inhibit human triple-negative breast cancer (MDA-MB-231) cells through induction of apoptosis and cell-cycle arrest. Ecological studies have suggested a possible association between ultraviolet B (UVB) radiation and reduction in the risk of breast cancer. However, the underlying mechanism of the combination of CoQ0 and UVB in human estrogen receptor–positive breast cancer (MCF-7) remains unclear. In this study, the possible effect of CoQ0 on inducing apoptosis in MCF-7 cells under exposure to low-dose UVB (0.05 J/cm2) has been investigated. CoQ0 treatment (0-35 µM, for 24-72 hours) inhibits moderately the growth of breast cancer MCF-7 cells, and the cell viability was significantly decreased when the cells were pretreated with UVB irradiation. It was noted that there was a remarkable accumulation of subploid cells, the so-called sub-G1 peak, in CoQ0-treated cells by using flow cytometric analysis, which suggests that the viability reduction observed after treatment may result from apoptosis induction in MCF-7 cells. CoQ0 caused an elevation of reactive oxygen species, as indicated by dichlorofluorescein fluorescence, and UVB pretreatment significantly increased CoQ0-induced reactive oxygen species generation in MCF-7 cells. In addition, cells were exposed to CoQ0, and the induction of DNA damage was evaluated by single-cell gel electrophoresis (comet assay). CoQ0-induced DNA damage was remarkably enhanced by UVB pretreatment. Furthermore, CoQ0 induced apoptosis in MCF-7 cells, which was associated with PARP degradation, Bcl-2/Bax dysregulation, and p53 expression as shown by western blot. Collectively, these findings suggest that CoQ0 might be an important supplemental agent for treating patients with breast cancer.
Collapse
Affiliation(s)
- Hui-Min Wang
- 1 National Chung Hsing University, Taichung, Taiwan
| | | | | | | | | | | | - Jer-Yuh Liu
- 2 China Medical University, Taichung, Taiwan
| | - Li-Sung Hsu
- 5 Chung Shan Medical University, Taichung, Taiwan
| | - Hsueh-Wei Chang
- 1 National Chung Hsing University, Taichung, Taiwan.,6 Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - You-Cheng Hseu
- 2 China Medical University, Taichung, Taiwan.,3 Asia University, Taichung, Taiwan
| |
Collapse
|