1
|
Rojas-Solé C, Pinilla-González V, Lillo-Moya J, González-Fernández T, Saso L, Rodrigo R. Integrated approach to reducing polypharmacy in older people: exploring the role of oxidative stress and antioxidant potential therapy. Redox Rep 2024; 29:2289740. [PMID: 38108325 PMCID: PMC10732214 DOI: 10.1080/13510002.2023.2289740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Increased life expectancy, attributed to improved access to healthcare and drug development, has led to an increase in multimorbidity, a key contributor to polypharmacy. Polypharmacy is characterised by its association with a variety of adverse events in the older persons. The mechanisms involved in the development of age-related chronic diseases are largely unknown; however, altered redox homeostasis due to ageing is one of the main theories. In this context, the present review explores the development and interaction between different age-related diseases, mainly linked by oxidative stress. In addition, drug interactions in the treatment of various diseases are described, emphasising that the holistic management of older people and their pathologies should prevail over the individual treatment of each condition.
Collapse
Affiliation(s)
- Catalina Rojas-Solé
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Víctor Pinilla-González
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - José Lillo-Moya
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Tommy González-Fernández
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, Rome, Italy
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
2
|
Stähli A, De Ry SP, Roccuzzo A, Imber JC, Sculean A. Effect of Coenzyme Q10 on early wound healing after recession coverage surgery with the modified coronally advanced tunnel technique and a connective tissue graft: A 6-month, triple-blinded, randomized, placebo-controlled pilot trial. Clin Oral Investig 2024; 28:424. [PMID: 38990401 PMCID: PMC11239743 DOI: 10.1007/s00784-024-05790-4] [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: 08/15/2023] [Accepted: 06/16/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVES Coenzyme Q10 (CoQ10) or ubiquinone is one of a cell's most important electron carriers during oxidative phosphorylation and many other cellular processes. As a strong anti-oxidant with further anti-inflammatory effects CoQ10 is of potential therapeutical value. The aim of this randomized controlled clinical trial was to investigate the effect of topical CoQ10 on early wound healing after recession coverage surgery using the modified coronally advanced tunnel (MCAT) and palatal connective tissue graft (CTG). MATERIALS AND METHODS Thirty patients with buccal gingival recessions were evaluated after being randomly allocated to: 1) MCAT and CTG with topical application of a coenzyme Q10 spray for 21 days or 2) MCAT and CTG with placebo spray. Wound healing was evaluated by the early wound healing index (EHI). Patient-reported pain was analyzed by a 100-mm visual analogue scale (VAS) at day 2, 7, 14 and 21 post-surgically. Mean recession coverage, gain of keratinized tissue and esthetic outcomes were assessed at 6 months. RESULTS EHI and pain scores showed no significant differences. Time to recovery defined as VAS<10 mm was shorter in the test group. Mean root coverage after 6 months was 84.62 ± 26.57% and 72.19 ± 26.30% for test and placebo, p=0.052. Complete root coverage was obtained in 9 (60%) test and in 2 (13.3%) placebo patients. Increase in keratinized tissue width and esthetical outcomes were similar for both groups. CONCLUSION CoQ10 had no significant effect on early wound healing and on mean root coverage after 6 months. CLINICAL RELEVANCE Early wound healing: in young healthy patients with no inflammatory oral conditions topical CoQ10 does not improve early healing.
Collapse
Affiliation(s)
- Alexandra Stähli
- Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, CH-3010, Berne, Switzerland.
| | - Siro P De Ry
- Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, CH-3010, Berne, Switzerland
| | - Andrea Roccuzzo
- Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, CH-3010, Berne, Switzerland
| | - Jean-Claude Imber
- Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, CH-3010, Berne, Switzerland
| | - Anton Sculean
- Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, CH-3010, Berne, Switzerland
| |
Collapse
|
3
|
Srivastava P, Bhoumik S, Yadawa AK, Kesherwani R, Rizvi SI. Coenzyme Q 10 supplementation affects cellular ionic balance: relevance to aging. Z NATURFORSCH C 2024; 0:znc-2024-0129. [PMID: 38963236 DOI: 10.1515/znc-2024-0129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 06/20/2024] [Indexed: 07/05/2024]
Abstract
Aging results into disruptive physiological functioning and cellular processes that affect the composition and structure of the plasma membrane. The plasma membrane is the major regulator of ionic homeostasis that regulates the functioning of membrane transporters and exchangers. Coenzyme Q10 is a lipid-soluble antioxidant molecule that declines during aging and age-associated diseases. The present study aims to explore the role of Coenzyme Q10 supplementation to rats during aging on membrane transporters and redox biomarkers. The study was conducted on young and old male Wistar rats supplemented with 20 mg/kg b.w. of Coenzyme Q10 per day. After a period of 28 days, rats were sacrificed and erythrocyte membrane was isolated. The result exhibits significant decline in biomarkers of oxidative stress in old control rats when compared with young control. The effect of Coenzyme Q10 supplementation was more pronounced in old rats. The functioning of membrane transporters and Na+/H+ exchanger showed potential return to normal levels in the Coenzyme Q10 treated rats. Overall, the results demonstrate that Coenzyme Q10 plays an important role in maintaining redox balance in cells which interconnects with membrane integrity. Thus, Coenzyme Q10 supplementation may play an important role in protecting age related alterations in erythrocyte membrane physiology.
Collapse
Affiliation(s)
- Parisha Srivastava
- Department of Biochemistry, 314956 University of Allahabad , Allahabad, Uttar Pradesh 211002, India
| | - Sukanya Bhoumik
- Department of Biochemistry, 314956 University of Allahabad , Allahabad, Uttar Pradesh 211002, India
| | - Arun K Yadawa
- Department of Biochemistry, 314956 University of Allahabad , Allahabad, Uttar Pradesh 211002, India
| | - Rashmi Kesherwani
- Department of Biochemistry, 314956 University of Allahabad , Allahabad, Uttar Pradesh 211002, India
| | - Syed Ibrahim Rizvi
- Department of Biochemistry, 314956 University of Allahabad , Allahabad, Uttar Pradesh 211002, India
| |
Collapse
|
4
|
Wang L, Wei Y, Sun Z, Tai W, Li H, Yin Y, Jiang LH, Wang JZ. Effectiveness and mechanisms of combined use of antioxidant nutrients in protecting against oxidative stress-induced neuronal loss and related neurological deficits. CNS Neurosci Ther 2024; 30:e14886. [PMID: 39072940 DOI: 10.1111/cns.14886] [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: 01/02/2024] [Revised: 06/04/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Oxidative stress is a well-known pathological factor driving neuronal loss and age-related neurodegenerative diseases. Melatonin, coenzyme Q10 and lecithin are three common nutrients with an antioxidative capacity. Here, we examined the effectiveness of them administrated individually and in combination in protecting against oxidative stress-induced neuronal death in vitro, and neurodegenerative conditions such as Alzheimer's disease and associated deficits in vivo. METHODS Mouse neuroblastoma Neuro-2a (N2a) cells were exposed with H2O2 for 6 h, and subsequently treated with melatonin, coenzyme Q10, and lecithin alone or in combination for further 24 h. Cell viability was assessed using the CCK-8 assay. Eight-week-old male mice were intraperitoneally injected with D-(+)-galactose for 10 weeks and administrated with melatonin, coenzyme Q10, lecithin, or in combination for 5 weeks starting from the sixth week, followed by behavioral tests to assess the effectiveness in mitigating neurological deficits, and biochemical assays to explore the underlying mechanisms. RESULTS Exposure to H2O2 significantly reduced the viability of N2a cells and increased oxidative stress and tau phosphorylation, all of which were alleviated by treatment with melatonin, coenzyme Q10, lecithin alone, and, most noticeably, by combined treatment. Administration of mice with D-(+)-galactose-induced oxidative stress and tau phosphorylation, brain aging, impairments in learning and memory, anxiety- and depression-like behaviors, and such detrimental effects were mitigated by melatonin, coenzyme Q10, lecithin alone, and, most consistently, by combined treatment. CONCLUSIONS These results suggest that targeting oxidative stress via supplementation of antioxidant nutrients, particularly in combination, is a better strategy to alleviate oxidative stress-mediated neuronal loss and brain dysfunction due to age-related neurodegenerative conditions.
Collapse
Affiliation(s)
- Lu Wang
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yingjuan Wei
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Department of Blood Transfusion, Xuchang Central Hospital, Xuchang, China
| | - Zhenzhou Sun
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Wenya Tai
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Hui Li
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yaling Yin
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Lin-Hua Jiang
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- School of Biomedical Sciences, University of Leeds, Leeds, UK
- EA4245, Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, Tours, France
| | - Jian-Zhi Wang
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Poljšak B, Milisav I. Decreasing Intracellular Entropy by Increasing Mitochondrial Efficiency and Reducing ROS Formation-The Effect on the Ageing Process and Age-Related Damage. Int J Mol Sci 2024; 25:6321. [PMID: 38928027 PMCID: PMC11203720 DOI: 10.3390/ijms25126321] [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: 04/23/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
A hypothesis is presented to explain how the ageing process might be influenced by optimizing mitochondrial efficiency to reduce intracellular entropy. Research-based quantifications of entropy are scarce. Non-equilibrium metabolic reactions and compartmentalization were found to contribute most to lowering entropy in the cells. Like the cells, mitochondria are thermodynamically open systems exchanging matter and energy with their surroundings-the rest of the cell. Based on the calculations from cancer cells, glycolysis was reported to produce less entropy than mitochondrial oxidative phosphorylation. However, these estimations depended on the CO2 concentration so that at slightly increased CO2, it was oxidative phosphorylation that produced less entropy. Also, the thermodynamic efficiency of mitochondrial respiratory complexes varies depending on the respiratory state and oxidant/antioxidant balance. Therefore, in spite of long-standing theoretical and practical efforts, more measurements, also in isolated mitochondria, with intact and suboptimal respiration, are needed to resolve the issue. Entropy increases in ageing while mitochondrial efficiency of energy conversion, quality control, and turnover mechanisms deteriorate. Optimally functioning mitochondria are necessary to meet energy demands for cellular defence and repair processes to attenuate ageing. The intuitive approach of simply supplying more metabolic fuels (more nutrients) often has the opposite effect, namely a decrease in energy production in the case of nutrient overload. Excessive nutrient intake and obesity accelerate ageing, while calorie restriction without malnutrition can prolong life. Balanced nutrient intake adapted to needs/activity-based high ATP requirement increases mitochondrial respiratory efficiency and leads to multiple alterations in gene expression and metabolic adaptations. Therefore, rather than overfeeding, it is necessary to fine-tune energy production by optimizing mitochondrial function and reducing oxidative stress; the evidence is discussed in this paper.
Collapse
Affiliation(s)
- Borut Poljšak
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia;
| | - Irina Milisav
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia;
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
6
|
Elgizawy EI, Amer GS, Ali EA, Alqalashy FS, Ibrahim MM, Latif AAA, Shaban AM. Comparing the efficacy of concomitant treatment of resistance exercise and creatine monohydrate versus multiple individual therapies in age related sarcopenia. Sci Rep 2024; 14:9798. [PMID: 38684784 PMCID: PMC11058861 DOI: 10.1038/s41598-024-59884-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/16/2024] [Indexed: 05/02/2024] Open
Abstract
Aging-related sarcopenia is a degenerative loss of strength and skeletal muscle mass that impairs quality of life. Evaluating NUDT3 gene and myogenin expression as new diagnostic tools in sarcopenia. Also, comparing the concomitant treatment of resistance exercise (EX) and creatine monohydrate (CrM) versus single therapy by EX, coenzyme Q10 (CoQ10), and CrM using aged rats. Sixty male rats were equally divided into groups. The control group, aging group, EX-treated group, the CoQ10 group were administered (500 mg/kg) of CoQ10, the CrM group supplied (0.3 mg/kg of CrM), and a group of CrM concomitant with resistance exercise. Serum lipid profiles, certain antioxidant markers, electromyography (EMG), nudix hydrolase 3 (NUDT3) expression, creatine kinase (CK), and sarcopenic index markers were measured after 12 weeks. The gastrocnemius muscle was stained with hematoxylin-eosin (H&E) and myogenin. The EX-CrM combination showed significant improvement in serum lipid profile, antioxidant markers, EMG, NUDT3 gene, myogenin expression, CK, and sarcopenic index markers from other groups. The NUDT3 gene and myogenin expression have proven efficient as diagnostic tools for sarcopenia. Concomitant treatment of CrM and EX is preferable to individual therapy because it reduces inflammation, improves the lipid serum profile, promotes muscle regeneration, and thus has the potential to improve sarcopenia.
Collapse
Affiliation(s)
- Eman I Elgizawy
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Yassin Abd El Ghafar St., Shebin El Kom, Menoufia, 32511, Egypt.
| | - Ghada S Amer
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Yassin Abd El Ghafar St., Shebin El Kom, Menoufia, 32511, Egypt
| | - Eman A Ali
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt
| | - Fatma S Alqalashy
- Pathology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt
| | - Marwa M Ibrahim
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt
| | - Asmaa A Abdel Latif
- Public Health and Community Medicine Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt
| | - Anwar M Shaban
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Yassin Abd El Ghafar St., Shebin El Kom, Menoufia, 32511, Egypt
| |
Collapse
|
7
|
Song YH, Lei HX, Yu D, Zhu H, Hao MZ, Cui RH, Meng XS, Sheng XH, Zhang L. Endogenous chemicals guard health through inhibiting ferroptotic cell death. Biofactors 2024; 50:266-293. [PMID: 38059412 DOI: 10.1002/biof.2015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 10/17/2023] [Indexed: 12/08/2023]
Abstract
Ferroptosis is a new form of regulated cell death caused by iron-dependent accumulation of lethal polyunsaturated phospholipids peroxidation. It has received considerable attention owing to its putative involvement in a wide range of pathophysiological processes such as organ injury, cardiac ischemia/reperfusion, degenerative disease and its prevalence in plants, invertebrates, yeasts, bacteria, and archaea. To counter ferroptosis, living organisms have evolved a myriad of intrinsic efficient defense systems, such as cyst(e)ine-glutathione-glutathione peroxidase 4 system (cyst(e)ine-GPX4 system), guanosine triphosphate cyclohydrolase 1/tetrahydrobiopterin (BH4) system (GCH1/BH4 system), ferroptosis suppressor protein 1/coenzyme Q10 system (FSP1/CoQ10 system), and so forth. Among these, GPX4 serves as the only enzymatic protection system through the reduction of lipid hydroperoxides, while other defense systems ultimately rely on small compounds to scavenge lipid radicals and prevent ferroptotic cell death. In this article, we systematically summarize the chemical biology of lipid radical trapping process by endogenous chemicals, such as coenzyme Q10 (CoQ10), BH4, hydropersulfides, vitamin K, vitamin E, 7-dehydrocholesterol, with the aim of guiding the discovery of novel ferroptosis inhibitors.
Collapse
Affiliation(s)
- Yuan-Hao Song
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, China
| | - Hong-Xu Lei
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
- Department of Chemistry, University of Chinese Academy of Sciences, Beijing, China
| | - Dou Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Hao Zhu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, China
| | - Meng-Zhu Hao
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, China
| | - Rong-Hua Cui
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, China
| | - Xiang-Shuai Meng
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, China
| | - Xie-Huang Sheng
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, China
| | - Lei Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Tissue Engineering Laboratory, Jinan, China
- Department of Radiology, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| |
Collapse
|
8
|
Zhang Q, Xia M, Zheng C, Yang Y, Bao J, Dai W, Mei X. The Cocrystal of Ubiquinol: Improved Stability and Bioavailability. Pharmaceutics 2023; 15:2499. [PMID: 37896258 PMCID: PMC10610044 DOI: 10.3390/pharmaceutics15102499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Coenzyme Q10 (CoQ10) exists in two forms, an oxidized form and a reduced form. Ubiquinol is the fully reduced form of CoQ10. Compared to the oxidized form, ubiquinol has a much higher biological absorption and better therapeutic effect. However, ubiquinol has an important stability problem which hampers its storage and formulation. It can be easily transformed into its oxidized form-ubiquinone-even at low temperature. In this work, we designed, synthesized, and characterized a new cocrystal of ubiquinol with vitamin B3 nicotinamide (UQ-NC). Compared to the marketed ubiquinol form, the cocrystal exhibited an excellent stability, improved dissolution properties, and higher bioavailability. The cocrystal remained stable for a long period, even when stored under stressed conditions. In the dissolution experiments, the cocrystal generated 12.6 (in SIF) and 38.3 (in SGF) times greater maximum ubiquinol concentrations above that of the marketed form. In addition, in the PK studies, compared to the marketed form, the cocrystal exhibited a 2.2 times greater maximum total coenzyme Q10 concentration and a 4.5 times greater AUC than that of the marketed form.
Collapse
Affiliation(s)
- Qi Zhang
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (Q.Z.); (M.X.); (C.Z.); (Y.Y.); (J.B.); (W.D.)
| | - Mengyuan Xia
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (Q.Z.); (M.X.); (C.Z.); (Y.Y.); (J.B.); (W.D.)
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Chenxuan Zheng
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (Q.Z.); (M.X.); (C.Z.); (Y.Y.); (J.B.); (W.D.)
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- College of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Yinghong Yang
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (Q.Z.); (M.X.); (C.Z.); (Y.Y.); (J.B.); (W.D.)
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Junjie Bao
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (Q.Z.); (M.X.); (C.Z.); (Y.Y.); (J.B.); (W.D.)
| | - Wenjuan Dai
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (Q.Z.); (M.X.); (C.Z.); (Y.Y.); (J.B.); (W.D.)
| | - Xuefeng Mei
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (Q.Z.); (M.X.); (C.Z.); (Y.Y.); (J.B.); (W.D.)
| |
Collapse
|
9
|
Kasai S, Kokubu D, Mizukami H, Itoh K. Mitochondrial Reactive Oxygen Species, Insulin Resistance, and Nrf2-Mediated Oxidative Stress Response-Toward an Actionable Strategy for Anti-Aging. Biomolecules 2023; 13:1544. [PMID: 37892226 PMCID: PMC10605809 DOI: 10.3390/biom13101544] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/12/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Reactive oxygen species (ROS) are produced mainly by mitochondrial respiration and function as signaling molecules in the physiological range. However, ROS production is also associated with the pathogenesis of various diseases, including insulin resistance (IR) and type 2 diabetes (T2D). This review focuses on the etiology of IR and early events, especially mitochondrial ROS (mtROS) production in insulin-sensitive tissues. Importantly, IR and/or defective adipogenesis in the white adipose tissues (WAT) is thought to increase free fatty acid and ectopic lipid deposition to develop into systemic IR. Fatty acid and ceramide accumulation mediate coenzyme Q reduction and mtROS production in IR in the skeletal muscle, while coenzyme Q synthesis downregulation is also involved in mtROS production in the WAT. Obesity-related IR is associated with the downregulation of mitochondrial catabolism of branched-chain amino acids (BCAAs) in the WAT, and the accumulation of BCAA and its metabolites as biomarkers in the blood could reliably indicate future T2D. Transcription factor NF-E2-related factor 2 (Nrf2), which regulates antioxidant enzyme expression in response to oxidative stress, is downregulated in insulin-resistant tissues. However, Nrf2 inducers, such as sulforaphane, could restore Nrf2 and target gene expression and attenuate IR in multiple tissues, including the WAT.
Collapse
Affiliation(s)
- Shuya Kasai
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| | - Daichi Kokubu
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
- Diet & Well-being Research Institute, KAGOME CO., LTD., 17 Nishitomiyama, Nasushiobara 329-2762, Japan
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| | - Ken Itoh
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| |
Collapse
|
10
|
Rebelo AP, Tomaselli PJ, Medina J, Wang Y, Dohrn MF, Nyvltova E, Danzi MC, Garrett M, Smith SE, Pestronk A, Li C, Ruiz A, Jacobs E, Feely SME, França MC, Gomes MV, Santos DF, Kumar S, Lombard DB, Saporta M, Hekimi S, Barrientos A, Weihl C, Shy ME, Marques W, Zuchner S. Biallelic variants in COQ7 cause distal hereditary motor neuropathy with upper motor neuron signs. Brain 2023; 146:4191-4199. [PMID: 37170631 PMCID: PMC10545612 DOI: 10.1093/brain/awad158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 04/12/2023] [Accepted: 04/23/2023] [Indexed: 05/13/2023] Open
Abstract
COQ7 encodes a hydroxylase responsible for the penultimate step of coenzyme Q10 (CoQ10) biosynthesis in mitochondria. CoQ10 is essential for multiple cellular functions, including mitochondrial oxidative phosphorylation, lipid metabolism, and reactive oxygen species homeostasis. Mutations in COQ7 have been previously associated with primary CoQ10 deficiency, a clinically heterogeneous multisystemic mitochondrial disorder. We identified COQ7 biallelic variants in nine families diagnosed with distal hereditary motor neuropathy with upper neuron involvement, expending the clinical phenotype associated with defects in this gene. A recurrent p.Met1? change was identified in five families from Brazil with evidence of a founder effect. Fibroblasts isolated from patients revealed a substantial depletion of COQ7 protein levels, indicating protein instability leading to loss of enzyme function. High-performance liquid chromatography assay showed that fibroblasts from patients had reduced levels of CoQ10, and abnormal accumulation of the biosynthetic precursor DMQ10. Accordingly, fibroblasts from patients displayed significantly decreased oxygen consumption rates in patients, suggesting mitochondrial respiration deficiency. Induced pluripotent stem cell-derived motor neurons from patient fibroblasts showed significantly increased levels of extracellular neurofilament light protein, indicating axonal degeneration. Our findings indicate a molecular pathway involving CoQ10 biosynthesis deficiency and mitochondrial dysfunction in patients with distal hereditary motor neuropathy. Further studies will be important to evaluate the potential benefits of CoQ10 supplementation in the clinical outcome of the disease.
Collapse
Affiliation(s)
- Adriana P Rebelo
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Pedro J Tomaselli
- Department of Neurology, University of São Paulo, Ribeirão Preto, 14048-900, Brazil
| | - Jessica Medina
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Ying Wang
- Department of Biology, McGill University, Montreal, QC, H3A 1A1, Canada
| | - Maike F Dohrn
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen 52074, Germany
| | - Eva Nyvltova
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Matt C Danzi
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Mark Garrett
- Department of Neurology, Washington University, St. Louis, MO 63112, USA
| | - Sean E Smith
- Department of Neurology, Washington University, St. Louis, MO 63112, USA
| | - Alan Pestronk
- Department of Neurology, Washington University, St. Louis, MO 63112, USA
| | - Chengcheng Li
- Department of Neurology, Washington University, St. Louis, MO 63112, USA
| | - Ariel Ruiz
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Elizabeth Jacobs
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Shawna M E Feely
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Marcondes C França
- Department of Neurology, University of São Paulo, Ribeirão Preto, 14048-900, Brazil
| | - Marcus V Gomes
- Department of Neurology, University of São Paulo, Ribeirão Preto, 14048-900, Brazil
| | - Diogo F Santos
- Department of Neurology, Federal University of Uberlândia, Uberlândia, MG 38405-320, Brazil
| | - Surinder Kumar
- Department of Pathology & Laboratory Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David B Lombard
- Department of Pathology & Laboratory Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Mario Saporta
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Siegfried Hekimi
- Department of Biology, McGill University, Montreal, QC, H3A 1A1, Canada
| | - Antoni Barrientos
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Conrad Weihl
- Department of Neurology, Washington University, St. Louis, MO 63112, USA
| | - Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Wilson Marques
- Department of Neurology, University of São Paulo, Ribeirão Preto, 14048-900, Brazil
| | - Stephan Zuchner
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
11
|
Nie X, Dong X, Hu Y, Xu F, Hu C, Shu C. Coenzyme Q10 Stimulate Reproductive Vatality. Drug Des Devel Ther 2023; 17:2623-2637. [PMID: 37667786 PMCID: PMC10475284 DOI: 10.2147/dddt.s386974] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023] Open
Abstract
Female infertility and pregnancy maintenance are associate with various factors, including quantity and quality of oocytes, genital inflammation, endometriosis, and other diseases. Women are even diagnosed as unexplained infertility or unexplained recurrent spontaneous abortion when failed to achieve pregnancy with current treatment, which are urgent clinical issues need to be addressed. Coenzyme Q10 (CoQ10) is a lipid-soluble electron carrier in the mitochondrial electron transport chain. It is not only essential for the mitochondria to produce energy, but also function as an antioxidant to maintain redox homeostasis in the body. Recently, the capacity of CoQ10 to reduce oxidative stress (OS), enhance mitochondrial activity, regulate gene expression and inhibit inflammatory responses, has been discovered as a novel adjuvant in male reproductive performance enhancing in both animal and human studies. Furthermore, CoQ10 is also proved to regulate immune balance, antioxidant, promote glucose and lipid metabolism. These properties will bring highlight for ovarian dysfunction reversing, ovulation ameliorating, oocyte maturation/fertilization promoting, and embryonic development optimizing. In this review, we systematically discuss the pleiotropic effects of CoQ10 in female reproductive disorders to investigate the mechanism and therapeutic potential to provide a reference in subsequent studies.
Collapse
Affiliation(s)
- Xinyu Nie
- Obstetrics and Gynecology Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
- Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Xinru Dong
- Obstetrics and Gynecology Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
- Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Yuge Hu
- Obstetrics and Gynecology Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
- Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Fangjun Xu
- Obstetrics and Gynecology Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Cong Hu
- Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Chang Shu
- Obstetrics and Gynecology Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
12
|
López-Lluch G. Coenzyme Q-related compounds to maintain healthy mitochondria during aging. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:277-308. [PMID: 37437981 DOI: 10.1016/bs.apcsb.2023.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Mitochondrial dysfunction is one of the main factors that affects aging progression and many age-related diseases. Accumulation of dysfunctional mitochondria can be driven by unbalanced mito/autophagy or by decrease in mitochondrial biosynthesis and turnover. Coenzyme Q is an essential component of the mitochondrial electron transport chain and a key factor in the protection of membrane and mitochondrial DNA against oxidation. Coenzyme Q levels decay during aging and this can be considered an accelerating factor in mitochondrial dysfunction and aging progression. Supplementation with coenzyme Q is successful for some tissues and organs but not for others. For this reason, the role of coenzyme Q in systemic aging is a complex picture that needs different strategies depending on the organ considered the main objective to be addressed. In this chapter we focus on the different effects of coenzyme Q and related compounds and the probable strategies to induce endogenous synthesis to maintain healthy aging.
Collapse
Affiliation(s)
- Guillermo López-Lluch
- Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide, Sevilla, Spain.
| |
Collapse
|
13
|
El-Bassouny DR, Mansour AA, Ellakkany AS, Ayuob NN, AbdElfattah AA. Can coenzyme Q10 alleviate the toxic effect of fenofibrate on skeletal muscle? Histochem Cell Biol 2023:10.1007/s00418-023-02205-5. [PMID: 37270716 PMCID: PMC10386954 DOI: 10.1007/s00418-023-02205-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/05/2023]
Abstract
Fenofibrate (FEN) is an antilipidemic drug that increases the activity of the lipoprotein lipase enzyme, thus enhancing lipolysis; however, it may cause myopathy and rhabdomyolysis in humans. Coenzyme Q10 (CoQ10) is an endogenously synthesized compound that is found in most living cells and plays an important role in cellular metabolism. It acts as the electron carrier in the mitochondrial respiratory chain. This study aimed to elucidate FEN-induced skeletal muscle changes in rats and to evaluate CoQ10 efficacy in preventing or alleviating these changes. Forty adult male rats were divided equally into four groups: the negative control group that received saline, the positive control group that received CoQ10, the FEN-treated group that received FEN, and the FEN + CoQ10 group that received both FEN followed by CoQ10 daily for 4 weeks. Animals were sacrificed and blood samples were collected to assess creatine kinase (CK). Soleus muscle samples were taken and processed for light and electron microscopic studies. This study showed that FEN increased CK levels and induced inflammatory cellular infiltration and disorganization of muscular architecture with lost striations. FEN increased the percentage of degenerated collagen fibers and immune expression of caspase-3. Ultrastructurally, FEN caused degeneration of myofibrils with distorted cell organelles. Treatment with CoQ10 could markedly ameliorate these FEN-induced structural changes and mostly regain the normal architecture of muscle fibers due to its antifibrotic and antiapoptotic effects. In conclusion, treatment with CoQ10 improved muscular structure by suppressing oxidative stress, attenuating inflammation, and inhibiting apoptosis.
Collapse
Affiliation(s)
- Dalia R El-Bassouny
- Medical Histology & Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Alyaa A Mansour
- Medical Histology & Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amany S Ellakkany
- Medical Histology & Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nasra N Ayuob
- Medical Histology Department, Faculty of Medicine, Damietta University, Damietta, Egypt
- Yousef Abdullatif Jameel Chair of Prophetic Medical Applications (YAJCPMA), Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amany A AbdElfattah
- Medical Histology & Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
- Department of Basic Medical Sciences, Faculty of Medicine, King Salman International University, South Sinai, El-Tor, Egypt.
| |
Collapse
|
14
|
Cao S, Yan H, Tang W, Zhang H, Liu J. Effects of dietary coenzyme Q10 supplementation during gestation on the embryonic survival and reproductive performance of high-parity sows. J Anim Sci Biotechnol 2023; 14:75. [PMID: 37264441 DOI: 10.1186/s40104-023-00879-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/05/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND Fertility declines in high-parity sows. This study investigated whether parity-dependent declines in embryonic survival and reproductive performance could be restored by dietary coenzyme Q10 (CoQ10) supplementation. METHODS Two experiments were performed. In Exp. 1, 30 young sows that had completed their 2nd parity and 30 high-parity sows that had completed their 10th parity, were fed either a control diet (CON) or a CON diet supplemented with 1 g/kg CoQ10 (+ CoQ10) from mating until slaughter at day 28 of gestation. In Exp. 2, a total of 314 post-weaning sows with two to nine parities were fed the CON or + CoQ10 diets from mating throughout gestation. RESULTS In Exp. 1, both young and high-parity sows had a similar number of corpora lutea, but high-parity sows had lower plasma CoQ10 concentrations, down-regulated genes involved with de novo CoQ10 synthesis in the endometrium tissues, and greater levels of oxidative stress markers in plasma and endometrium tissues. High-parity sows had fewer total embryos and alive embryos, lower embryonic survival, and greater embryo mortality than young sows. Dietary CoQ10 supplementation increased the number of live embryos and the embryonic survival rate to levels similar to those of young sows, as well as lowering the levels of oxidative stress markers. In Exp. 2, sows showed a parity-dependent decline in plasma CoQ10 levels, and sows with more than four parities showed a progressive decline in the number of total births, live births, and piglets born effective. Dietary supplementation with CoQ10 increased the number of total births, live births, and born effective, and decreased the intra-litter covariation coefficients and the percentage of sows requiring farrowing assistance during parturition. CONCLUSIONS Dietary CoQ10 supplementation can improve the embryonic survival and reproductive performance of gestating sows with high parity, probably by improving the development of uterine function.
Collapse
Affiliation(s)
- Shanchuan Cao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, China
- Department of Animal Resource and Science, Dankook University, Cheonan, 31116, Korea
| | - Honglin Yan
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, China
| | - Wenjie Tang
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtech Feed Co., Ltd., Chengdu, 610066, China
| | - Hongfu Zhang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, China.
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, China.
| |
Collapse
|
15
|
Inoue R, Miura M, Yanai S, Nishimune H. Coenzyme Q 10 supplementation improves the motor function of middle-aged mice by restoring the neuronal activity of the motor cortex. Sci Rep 2023; 13:4323. [PMID: 36922562 PMCID: PMC10017826 DOI: 10.1038/s41598-023-31510-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Physiological aging causes motor function decline and anatomical and biochemical changes in the motor cortex. We confirmed that middle-aged mice at 15-18 months old show motor function decline, which can be restored to the young adult level by supplementing with mitochondrial electron transporter coenzyme Q10 (CoQ10) as a water-soluble nanoformula by drinking water for 1 week. CoQ10 supplementation concurrently improved brain mitochondrial respiration but not muscle strength. Notably, we identified an age-related decline in field excitatory postsynaptic potential (fEPSP) amplitude in the pathway from layers II/III to V of the primary motor area of middle-aged mice, which was restored to the young adult level by supplementing with CoQ10 for 1 week but not by administering CoQ10 acutely to brain slices. Interestingly, CoQ10 with high-frequency stimulation induced NMDA receptor-dependent long-term potentiation (LTP) in layer V of the primary motor cortex of middle-aged mice. Importantly, the fEPSP amplitude showed a larger input‒output relationship after CoQ10-dependent LTP expression. These data suggest that CoQ10 restores the motor function of middle-aged mice by improving brain mitochondrial function and the basal fEPSP level of the motor cortex, potentially by enhancing synaptic plasticity efficacy. Thus, CoQ10 supplementation may ameliorate the age-related decline in motor function in humans.
Collapse
Affiliation(s)
- Ritsuko Inoue
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan.
| | - Masami Miura
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan.,Saitama Central Hospital, 2177 Kamitome, Miyoshicho, Iruma-Gun, Saitama, 354-0045, Japan
| | - Shuichi Yanai
- Laboratory of Memory Neuroscience, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Hiroshi Nishimune
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan. .,Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-8-1 Harumicho, Fuchu-Shi, Tokyo, 183-8538, Japan.
| |
Collapse
|
16
|
García-López C, García-López V, Matamoros JA, Fernández-Albarral JA, Salobrar-García E, de Hoz R, López-Cuenca I, Sánchez-Puebla L, Ramírez JM, Ramírez AI, Salazar JJ. The Role of Citicoline and Coenzyme Q10 in Retinal Pathology. Int J Mol Sci 2023; 24:5072. [PMID: 36982157 PMCID: PMC10049438 DOI: 10.3390/ijms24065072] [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: 12/02/2022] [Revised: 02/16/2023] [Accepted: 03/05/2023] [Indexed: 03/09/2023] Open
Abstract
Ocular neurodegenerative diseases such as glaucoma, diabetic retinopathy, and age-related macular degeneration are common retinal diseases responsible for most of the blindness causes in the working-age and elderly populations in developed countries. Many of the current treatments used in these pathologies fail to stop or slow the progression of the disease. Therefore, other types of treatments with neuroprotective characteristics may be necessary to allow a more satisfactory management of the disease. Citicoline and coenzyme Q10 are molecules that have neuroprotective, antioxidant, and anti-inflammatory properties, and their use could have a beneficial effect in ocular neurodegenerative pathologies. This review provides a compilation, mainly from the last 10 years, of the main studies that have been published on the use of these drugs in these neurodegenerative diseases of the retina, analyzing the usefulness of these drugs in these pathologies.
Collapse
Affiliation(s)
- Claudia García-López
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Verónica García-López
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - José A. Matamoros
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - José A. Fernández-Albarral
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Elena Salobrar-García
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Inés López-Cuenca
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Lidia Sánchez-Puebla
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - José M. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Medicina, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Ana I. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Juan J. Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| |
Collapse
|
17
|
Sharma A, Chabloz S, Lapides RA, Roider E, Ewald CY. Potential Synergistic Supplementation of NAD+ Promoting Compounds as a Strategy for Increasing Healthspan. Nutrients 2023; 15:nu15020445. [PMID: 36678315 PMCID: PMC9861325 DOI: 10.3390/nu15020445] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Disrupted biological function, manifesting through the hallmarks of aging, poses one of the largest threats to healthspan and risk of disease development, such as metabolic disorders, cardiovascular ailments, and neurodegeneration. In recent years, numerous geroprotectors, senolytics, and other nutraceuticals have emerged as potential disruptors of aging and may be viable interventions in the immediate state of human longevity science. In this review, we focus on the decrease in nicotinamide adenine dinucleotide (NAD+) with age and the supplementation of NAD+ precursors, such as nicotinamide mononucleotide (NMN) or nicotinamide riboside (NR), in combination with other geroprotective compounds, to restore NAD+ levels present in youth. Furthermore, these geroprotectors may enhance the efficacy of NMN supplementation while concurrently providing their own numerous health benefits. By analyzing the prevention of NAD+ degradation through the inhibition of CD38 or supporting protective downstream agents of SIRT1, we provide a potential framework of the CD38/NAD+/SIRT1 axis through which geroprotectors may enhance the efficacy of NAD+ precursor supplementation and reduce the risk of age-related diseases, thereby potentiating healthspan in humans.
Collapse
Affiliation(s)
- Arastu Sharma
- Laboratory of Extracellular Matrix Regeneration, Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zürich, 8603 Schwerzenbach, Switzerland
- AVEA Life AG, Bahnhofplatz, 6300 Zug, Switzerland
| | | | - Rebecca A. Lapides
- Department of Dermatology, University Hospital of Basel, 4031 Basel, Switzerland
- Robert Larner, MD College of Medicine at the University of Vermont, Burlington, VT 05405, USA
| | - Elisabeth Roider
- Department of Dermatology, University Hospital of Basel, 4031 Basel, Switzerland
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
- Maximon AG, Bahnhofplatz, 6300 Zug, Switzerland
| | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zürich, 8603 Schwerzenbach, Switzerland
- Correspondence:
| |
Collapse
|
18
|
Chhitij T, Seo JE, Keum T, Noh G, Bashyal S, Lamichhane S, Kim JH, Lee JH, Park JH, Choi J, Song SH, Lee S. Optimized self-microemulsifying drug delivery system improves the oral bioavailability and brain delivery of coenzyme Q 10. Drug Deliv 2022; 29:2330-2342. [PMID: 35850616 PMCID: PMC9848412 DOI: 10.1080/10717544.2022.2100515] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Our study aimed to develop a self-microemulsifying drug delivery system for the poorly aqueous-soluble drug Coenzyme Q10, to improve the dissolution and the oral bioavailability. Excipients were selected based on their Coenzyme Q10 solubility, and their concentrations were set for the optimization of the microemulsion by using a D-optimal mixture design to achieve a minimum droplet size and a maximum solubility of Coenzyme Q10 within 15 min. The optimized formulation was composed of an oil (omega-3; 38.55%), a co-surfactant (Lauroglycol® 90; 31.42%), and a surfactant (Gelucire® 44/14; 30%) and exhibited a mean droplet size of 237.6 ± 5.8 nm and a drug solubilization (at 15 min) of 16 ± 2.48%. The drug dissolution of the optimized formulation conducted over 8 h in phosphate buffer medium (pH 6.8) was significantly higher when compared to that of the Coenzyme Q10 suspension. A pharmacokinetic study in rats revealed a 4.5-fold and a 4.1-fold increase in the area under curve and the peak plasma concentration values generated by the optimized formulation respectively, as compared to the Coenzyme Q10 suspension. A Coenzyme Q10 brain distribution study revealed a higher Coenzyme Q10 distribution in the brains of rats treated with the optimized formulation than the Coenzyme Q10 suspension. Coenzyme Q10-loaded self microemulsifying drug delivery system was successfully formulated and optimized by a response surface methodology based on a D-optimal mixture design and could be used as a delivery vehicle for the enhancement of the oral bioavailability and brain distribution of poorly soluble drugs such as Coenzyme Q10.
Collapse
Affiliation(s)
- Thapa Chhitij
- Center for Forensic Pharmaceutical Sciences, College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Jo-Eun Seo
- Center for Forensic Pharmaceutical Sciences, College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Taekwang Keum
- Center for Forensic Pharmaceutical Sciences, College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Gyubin Noh
- Center for Forensic Pharmaceutical Sciences, College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Santosh Bashyal
- Center for Forensic Pharmaceutical Sciences, College of Pharmacy, Keimyung University, Daegu, Republic of Korea,Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Shrawani Lamichhane
- Center for Forensic Pharmaceutical Sciences, College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Jung Hwan Kim
- Center for Forensic Pharmaceutical Sciences, College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Jae Heon Lee
- Center for Forensic Pharmaceutical Sciences, College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Jee Hun Park
- R&D Center, Korean Drug Co., Ltd, Seoul, Republic of Korea
| | - Jaewoong Choi
- Center for Forensic Pharmaceutical Sciences, College of Pharmacy, Keimyung University, Daegu, Republic of Korea,R&D Center, Korean Drug Co., Ltd, Seoul, Republic of Korea
| | - Se Hyun Song
- College of Pharmacy, Kyungsung University, Busan, 48434, Republic of Korea
| | - Sangkil Lee
- Center for Forensic Pharmaceutical Sciences, College of Pharmacy, Keimyung University, Daegu, Republic of Korea,Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA,CONTACT Sangkil Lee Center for Forensic Pharmaceutical Sciences, College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu42601, Republic of Korea, Tel: +82-53-580-6655, FAX: +82-53-580-5164
| |
Collapse
|
19
|
Gasmi A, Bjørklund G, Mujawdiya PK, Semenova Y, Piscopo S, Peana M. Coenzyme Q 10 in aging and disease. Crit Rev Food Sci Nutr 2022; 64:3907-3919. [PMID: 36300654 DOI: 10.1080/10408398.2022.2137724] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Coenzyme Q10 (CoQ10) is an essential component of the electron transport chain. It also acts as an antioxidant in cellular membranes. It can be endogenously produced in all cells by a specialized mitochondrial pathway. CoQ10 deficiency, which can result from aging or insufficient enzyme function, has been considered to increase oxidative stress. Some drugs, including statins and bisphosphonates, often used by older individuals, can interfere with enzymes responsible for endogenous CoQ10 synthesis. Oral supplementation with high doses of CoQ10 can increase both its circulating and intracellular levels and several clinical trials observed that its administration provided beneficial effects on different disorders such as cardiovascular disease and inflammation which have been associated with low CoQ10 levels and high oxidative stress. Moreover, CoQ10 has been suggested as a promising therapeutic agent to prevent and slow the progression of other diseases including metabolic syndrome and type 2 diabetes, neurodegenerative and male infertility. However, there is still a need for further studies and well-designed clinical trials involving a large number of participants undergoing longer treatments to assess the benefits of CoQ10 for these disorders.
Collapse
Affiliation(s)
- Amin Gasmi
- Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, Villeurbanne, France
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| | | | - Yuliya Semenova
- Nazarbayev University School of Medicine, Astana, Kazakhstan
| | - Salva Piscopo
- Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, Villeurbanne, France
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
20
|
Detrimental Effects of Lipid Peroxidation in Type 2 Diabetes: Exploring the Neutralizing Influence of Antioxidants. Antioxidants (Basel) 2022; 11:antiox11102071. [PMID: 36290794 PMCID: PMC9598619 DOI: 10.3390/antiox11102071] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Lipid peroxidation, including its prominent byproducts such as malondialdehyde (MDA) and 4-hydroxy-2-nonenal (4-HNE), has long been linked with worsened metabolic health in patients with type 2 diabetes (T2D). In fact, patients with T2D already display increased levels of lipids in circulation, including low-density lipoprotein-cholesterol and triglycerides, which are easily attacked by reactive oxygen molecules to give rise to lipid peroxidation. This process severely depletes intracellular antioxidants to cause excess generation of oxidative stress. This consequence mainly drives poor glycemic control and metabolic complications that are implicated in the development of cardiovascular disease. The current review explores the pathological relevance of elevated lipid peroxidation products in T2D, especially highlighting their potential role as biomarkers and therapeutic targets in disease severity. In addition, we briefly explain the implication of some prominent antioxidant enzymes/factors involved in the blockade of lipid peroxidation, including termination reactions that involve the effect of antioxidants, such as catalase, coenzyme Q10, glutathione peroxidase, and superoxide dismutase, as well as vitamins C and E.
Collapse
|
21
|
The Modulation of Ubiquinone, a Lipid Antioxidant, on Neuronal Voltage-Gated Sodium Current. Nutrients 2022; 14:nu14163393. [PMID: 36014898 PMCID: PMC9413396 DOI: 10.3390/nu14163393] [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: 07/09/2022] [Revised: 08/04/2022] [Accepted: 08/16/2022] [Indexed: 12/02/2022] Open
Abstract
Ubiquinone, composed of a 1,4-benzoquinone and naturally produced in the body, actively participates in the mitochondrial redox reaction and functions as an endogenous lipid antioxidant, protecting against peroxidation in the pituitary-dependent hormonal system. However, the questions of if and how ubiquinone directly affects neuronal ionic currents remain largely unsettled. We investigated its effects on ionic currents in pituitary neurons (GH3 and MMQ cells) with the aid of patch-clamp technology. Ubiquinone decreased the peak amplitude of the voltage-gated Na+ current (INa) with a slowing of the inactivation rate. Neither menadione nor superoxide dismutase modified the ubiquinone-induced INa inhibition. In response to an isosceles-triangular ramp pulse, the persistent INa (INa(P)) at high- and low- threshold potentials occurred concurrently with a figure-eight hysteresis loop. With ubiquinone, the INa(P) increased with no change in the intersection voltage, and the magnitude of the voltage-dependent hysteresis of the current was enhanced. Ubiquinone was ineffective in modifying the gating of hyperpolarization-activated cation currents. In MMQ lactotrophs, ubiquinone effectively decreased the amplitude of the INa and the current inactivation rate. In sum, the effects of ubiquinone demonstrated herein occur upstream of its effects on mitochondrial redox processes, involved in its modulation of sodium channels and neuronal excitability.
Collapse
|
22
|
Silva SVE, Gallia MC, da Luz JRD, de Rezende AA, Bongiovanni GA, Araujo-Silva G, Almeida MDG. Antioxidant Effect of Coenzyme Q10 in the Prevention of Oxidative Stress in Arsenic-Treated CHO-K1 Cells and Possible Participation of Zinc as a Pro-Oxidant Agent. Nutrients 2022; 14:nu14163265. [PMID: 36014770 PMCID: PMC9412518 DOI: 10.3390/nu14163265] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
Oxidative stress is an imbalance between levels of reactive oxygen species (ROS) and antioxidant enzymes. Compounds with antioxidant properties, such as coenzyme Q10 (CoQ10), can reduce cellular imbalance caused by an increase in ROS. CoQ10 participates in modulating redox homeostasis due to its antioxidant activity and its preserving mitochondrial functions. Thus, the present study demonstrated the protective effects of CoQ10 against oxidative stress and cytotoxicity induced by arsenic (As). Antioxidant capacity, formation of hydroperoxides, generation of ROS, and the effect on cellular viability of CoQ10, were investigated to determine the protective effect of CoQ10 against As and pro-oxidant compounds, such as zinc. Cell viability assays showed that CoQ10 is cytoprotective under cellular stress conditions, with potent antioxidant activity, regardless of the concentration tested. Zn, when used at higher concentrations, can increase ROS and show a pro-oxidant effect causing cell damage. The cytotoxic effect observed for As, Zn, or the combination of both could be prevented by CoQ10, without any decrease in its activity at cellular levels when combined with Zn.
Collapse
Affiliation(s)
- Saulo Victor e Silva
- Post-Graduation Program in Pharmaceutical Sciences, Multidisciplinary Research Laboratory, Department of Clinical and Toxicological Analysis (DACT), Health Sciences Center, Federal University of the Rio Grande do Norte (UFRN), Natal 59012570, Brazil
| | - María Celeste Gallia
- Institute of Research and Development in Process Engineering, Biotechnology and Alternative Energies (PROBIEN), National Council for Scientific and Technical Research (CONICET), School of Agricultural Sciences, Neuquén 8300, Argentina
| | - Jefferson Romáryo Duarte da Luz
- Post-Graduation Program in Health Sciences, Multidisciplinary Research Laboratory, Department of Clinical and Toxicological Analysis (DACT), Health Sciences Center, UFRN, Natal 59012570, Brazil
- Organic Chemistry and Biochemistry Laboratory, State University of Amapá (UEAP), Macapá 68900070, Brazil
| | - Adriana Augusto de Rezende
- Post-Graduation Program in Pharmaceutical Sciences, Multidisciplinary Research Laboratory, Department of Clinical and Toxicological Analysis (DACT), Health Sciences Center, Federal University of the Rio Grande do Norte (UFRN), Natal 59012570, Brazil
- Post-Graduation Program in Health Sciences, Multidisciplinary Research Laboratory, Department of Clinical and Toxicological Analysis (DACT), Health Sciences Center, UFRN, Natal 59012570, Brazil
| | - Guillermina Azucena Bongiovanni
- Institute of Research and Development in Process Engineering, Biotechnology and Alternative Energies (PROBIEN), National Council for Scientific and Technical Research (CONICET), School of Agricultural Sciences, Neuquén 8300, Argentina
| | - Gabriel Araujo-Silva
- Organic Chemistry and Biochemistry Laboratory, State University of Amapá (UEAP), Macapá 68900070, Brazil
| | - Maria das Graças Almeida
- Post-Graduation Program in Pharmaceutical Sciences, Multidisciplinary Research Laboratory, Department of Clinical and Toxicological Analysis (DACT), Health Sciences Center, Federal University of the Rio Grande do Norte (UFRN), Natal 59012570, Brazil
- Post-Graduation Program in Health Sciences, Multidisciplinary Research Laboratory, Department of Clinical and Toxicological Analysis (DACT), Health Sciences Center, UFRN, Natal 59012570, Brazil
- Sciences Center, UFRN, Natal 59012570, Brazil
- Correspondence:
| |
Collapse
|
23
|
Chang PS, Chou HH, Lai TJ, Yen CH, Pan JC, Lin PT. Investigation of coenzyme Q10 status, serum amyloid-β, and tau protein in patients with dementia. Front Aging Neurosci 2022; 14:910289. [PMID: 35959290 PMCID: PMC9358012 DOI: 10.3389/fnagi.2022.910289] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/06/2022] [Indexed: 11/14/2022] Open
Abstract
Objectives Dementia is an oxidative stress-related disease. Coenzyme Q10 is a nutrient that occurs naturally in the human body and acts as an antioxidant. The purpose of this study was to investigate the relationships of coenzyme Q10 status, biomarkers for dementia (amyloid β and tau protein), and antioxidant capacity in patients with dementia. Methods Eighty dementia patients aged ≥60 years and with a mini mental state examination (MMSE) score ≤ 26 were enrolled. The levels of coenzyme Q10, total antioxidant capacity (TAC), amyloid β, and tau protein were measured. Results A total of 73% of patients had a low coenzyme Q10 status. Patients with low coenzyme Q10 status had a significantly higher level of serum amyloid β-42 and amyloid β-42/40 ratio (p < 0.05). Coenzyme Q10 status was significantly correlated with the values of TAC, MMSE score, amyloid β-42, and amyloid β-42/40 ratio (p < 0.05) but not with tau protein. Additionally, a high proportion of moderate dementia patients were found to have low coenzyme Q10 status (p = 0.07). Conclusion Patients with dementia suffered from coenzyme Q10 deficiency, and the degree of deficiency was related to the level of amyloid-β and antioxidant capacity. Since adequate level of coenzyme Q10 may delay the progression of dementia, monitoring coenzyme Q10 status in patients with dementia is necessary.
Collapse
Affiliation(s)
- Po-Sheng Chang
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
- Graduate Program in Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - Hsi-Hsien Chou
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Neurology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Te-Jen Lai
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chi-Hua Yen
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Family and Community Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ji-Cyun Pan
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - Ping-Ting Lin
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
- *Correspondence: Ping-Ting Lin,
| |
Collapse
|
24
|
Zhang Y, Huang X, Liu N, Liu M, Sun C, Qi B, Sun K, Wei X, Ma Y, Zhu L. Discovering the Potential Value of Coenzyme Q10 in Oxidative Stress: Enlightenment From a Synthesis of Clinical Evidence Based on Various Population. Front Pharmacol 2022; 13:936233. [PMID: 35910386 PMCID: PMC9330130 DOI: 10.3389/fphar.2022.936233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/09/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Oxidative stress (OS) is associated with ferroptosis. Coenzyme Q10 (CoQ10), as an adjuvant treatment, has shown to be beneficial against OS. However, the efficacy of CoQ10 as a therapeutic agent against OS has not been promptly updated and systematically investigated. Methods: A systematic literature search was performed using the Medline, EMBASE, Web of science, Cochrane Central Register of Controlled Trials, CNKI, CBM, Science direct and clinical trial. gov to identify randomized clinical trials evaluating the efficacy of CoQ10 supplementation on OS parameters. Standard mean differences and 95% confidence intervals were calculated for net changes in OS parameters using a random-effects model. Results: Twenty-one randomized clinical studies met the eligibility criteria to be included in the meta-analysis. Overall, CoQ10 supplementation increased the levels of antioxidant enzymes [including superoxide dismutase (SOD) (SMD = 0.63; 95% CI: 0.38 to 0.88; p < 0.001), catalase (CAT) (SMD = 0.44; 95% CI:0.16 to 0.72; p = 0.002)] significantly and the levels of malondialdehyde (MDA) (SMD = -0.68; 95% CI: 0.93 to -0.43; p < 0.001) was decreased considerably. However, significant associations were not observed between this supplement and total antioxidant capacity (TAC), glutathione peroxidase (GPx) activity. Conclusion: CoQ10 can improve OS as indicated by statistical significance in CAT and MDA concentrations, as well as SOD activity. Future studies focusing on long-term results and specific valuation of OS parameters are required to confirm the efficacy of CoQ10 on OS. We also believe that with the further research on ferroptosis, CoQ10 will gain more attention. Systematic Review Registration: [https://inplasy.com/], identifier [INPLASY2021120123].
Collapse
Affiliation(s)
- Yili Zhang
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinyi Huang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ning Liu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mengmin Liu
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chuanrui Sun
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baoyu Qi
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kai Sun
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xu Wei
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Xu Wei, ; Yong Ma, ; Liguo Zhu,
| | - Yong Ma
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Xu Wei, ; Yong Ma, ; Liguo Zhu,
| | - Liguo Zhu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Xu Wei, ; Yong Ma, ; Liguo Zhu,
| |
Collapse
|
25
|
Barcena ML, Aslam M, Pozdniakova S, Norman K, Ladilov Y. Cardiovascular Inflammaging: Mechanisms and Translational Aspects. Cells 2022; 11:cells11061010. [PMID: 35326461 PMCID: PMC8946971 DOI: 10.3390/cells11061010] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/07/2022] [Accepted: 03/15/2022] [Indexed: 12/14/2022] Open
Abstract
Aging is one of the major non-reversible risk factors for several chronic diseases, including cancer, type 2 diabetes, dementia, and cardiovascular diseases (CVD), and it is a key cause of multimorbidity, disability, and frailty (decreased physical activity, fatigue, and weight loss). The underlying cellular mechanisms are complex and consist of multifactorial processes, such as telomere shortening, chronic low-grade inflammation, oxidative stress, mitochondrial dysfunction, accumulation of senescent cells, and reduced autophagy. In this review, we focused on the molecular mechanisms and translational aspects of cardiovascular aging-related inflammation, i.e., inflammaging.
Collapse
Affiliation(s)
- Maria Luisa Barcena
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-525-359
| | - Muhammad Aslam
- Experimental Cardiology, Department of Internal Medicine I, Justus Liebig University, Aulweg 129, 35392 Giessen, Germany;
- Department of Cardiology, Kerckhoff Clinic GmbH, 61231 Bad Nauheim, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Rhein-Main, 61231 Bad Nauheim, Germany
| | - Sofya Pozdniakova
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- Barcelona Biomedical Research Park (PRBB), Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003 Barcelona, Spain
| | - Kristina Norman
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- Department of Nutrition & Gerontology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Yury Ladilov
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- Department of Cardiovascular Surgery, Heart Center Brandenburg, Brandenburg Medical School Theodor Fontane, University Hospital, Ladeburger Str. 17, 16321 Bernau, Germany
| |
Collapse
|
26
|
DiNicolantonio JJ, McCarty MF, O'Keefe JH. Coenzyme Q10 deficiency can be expected to compromise Sirt1 activity. Open Heart 2022; 9:e001927. [PMID: 35296520 PMCID: PMC8928362 DOI: 10.1136/openhrt-2021-001927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
For reasons that remain unclear, endogenous synthesis and tissue levels of coenzyme Q10 (CoQ10) tend to decline with increasing age in at least some tissues. When CoQ10 levels are sufficiently low, this compromises the efficiency of the mitochondrial electron transport chain, such that production of superoxide by site 2 increases and the rate of adenosine triphosphate production declines. Moreover, CoQ10 deficiency can be expected to decrease activities of Sirt1 and Sirt3 deacetylases, believed to be key determinants of health span. Reduction of the cytoplasmic and mitochondrial NAD+/NADH ratio consequent to CoQ10 deficit can be expected to decrease the activity of these deacetylases by lessening availability of their obligate substrate NAD+ The increased oxidant production induced by CoQ10 deficiency can decrease the stability of Sirt1 protein by complementary mechanisms. And CoQ10 deficiency has also been found to lower mRNA expression of Sirt1. An analysis of the roles of Sirt1/Sirt3 in modulation of cellular function helps to rationalise clinical benefits of CoQ10 supplementation reported in heart failure, hypertension, non-alcoholic fatty liver disease, metabolic syndrome and periodontal disease. Hence, correction of CoQ10 deficiency joins a growing list of measures that have potential for amplifying health protective Sirt1/Sirt3 activities.
Collapse
Affiliation(s)
- James J DiNicolantonio
- Department of Preventive Cardiology, Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA
| | | | - James H O'Keefe
- Saint Luke's Mid America Heart Institute, University of Missouri-Kansas City, Kansas City, Missouri, USA
| |
Collapse
|
27
|
Coenzyme Q at the Hinge of Health and Metabolic Diseases. Antioxidants (Basel) 2021; 10:antiox10111785. [PMID: 34829656 PMCID: PMC8615162 DOI: 10.3390/antiox10111785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/28/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022] Open
Abstract
Coenzyme Q is a unique lipidic molecule highly conserved in evolution and essential to maintaining aerobic metabolism. It is endogenously synthesized in all cells by a very complex pathway involving a group of nuclear genes that share high homology among species. This pathway is tightly regulated at transcription and translation, but also by environment and energy requirements. Here, we review how coenzyme Q reacts within mitochondria to promote ATP synthesis and also integrates a plethora of metabolic pathways and regulates mitochondrial oxidative stress. Coenzyme Q is also located in all cellular membranes and plasma lipoproteins in which it exerts antioxidant function, and its reaction with different extramitochondrial oxidoreductases contributes to regulate the cellular redox homeostasis and cytosolic oxidative stress, providing a key factor in controlling various apoptosis mechanisms. Coenzyme Q levels can be decreased in humans by defects in the biosynthesis pathway or by mitochondrial or cytosolic dysfunctions, leading to a highly heterogeneous group of mitochondrial diseases included in the coenzyme Q deficiency syndrome. We also review the importance of coenzyme Q levels and its reactions involved in aging and age-associated metabolic disorders, and how the strategy of its supplementation has had benefits for combating these diseases and for physical performance in aging.
Collapse
|
28
|
González-García P, Barriocanal-Casado E, Díaz-Casado ME, López-Herrador S, Hidalgo-Gutiérrez A, López LC. Animal Models of Coenzyme Q Deficiency: Mechanistic and Translational Learnings. Antioxidants (Basel) 2021; 10:antiox10111687. [PMID: 34829558 PMCID: PMC8614664 DOI: 10.3390/antiox10111687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/16/2022] Open
Abstract
Coenzyme Q (CoQ) is a vital lipophilic molecule that is endogenously synthesized in the mitochondria of each cell. The CoQ biosynthetic pathway is complex and not completely characterized, and it involves at least thirteen catalytic and regulatory proteins. Once it is synthesized, CoQ exerts a wide variety of mitochondrial and extramitochondrial functions thank to its redox capacity and its lipophilicity. Thus, low levels of CoQ cause diseases with heterogeneous clinical symptoms, which are not always understood. The decreased levels of CoQ may be primary caused by defects in the CoQ biosynthetic pathway or secondarily associated with other diseases. In both cases, the pathomechanisms are related to the CoQ functions, although further experimental evidence is required to establish this association. The conventional treatment for CoQ deficiencies is the high doses of oral CoQ10 supplementation, but this therapy is not effective for some specific clinical presentations, especially in those involving the nervous system. To better understand the CoQ biosynthetic pathway, the biological functions linked to CoQ and the pathomechanisms of CoQ deficiencies, and to improve the therapeutic outcomes of this syndrome, a variety of animal models have been generated and characterized in the last decade. In this review, we show all the animal models available, remarking on the most important outcomes that each model has provided. Finally, we also comment some gaps and future research directions related to CoQ metabolism and how the current and novel animal models may help in the development of future research studies.
Collapse
Affiliation(s)
- Pilar González-García
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
- Correspondence: (P.G.-G.); (L.C.L.)
| | - Eliana Barriocanal-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - María Elena Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Sergio López-Herrador
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Agustín Hidalgo-Gutiérrez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Luis C. López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
- Correspondence: (P.G.-G.); (L.C.L.)
| |
Collapse
|
29
|
Bilbao-Malavé V, González-Zamora J, de la Puente M, Recalde S, Fernandez-Robredo P, Hernandez M, Layana AG, Saenz de Viteri M. Mitochondrial Dysfunction and Endoplasmic Reticulum Stress in Age Related Macular Degeneration, Role in Pathophysiology, and Possible New Therapeutic Strategies. Antioxidants (Basel) 2021; 10:1170. [PMID: 34439418 PMCID: PMC8388889 DOI: 10.3390/antiox10081170] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
Age related macular degeneration (AMD) is the main cause of legal blindness in developed countries. It is a multifactorial disease in which a combination of genetic and environmental factors contributes to increased risk of developing this vision-incapacitating condition. Oxidative stress plays a central role in the pathophysiology of AMD and recent publications have highlighted the importance of mitochondrial dysfunction and endoplasmic reticulum stress in this disease. Although treatment with vascular endothelium growth factor inhibitors have decreased the risk of blindness in patients with the exudative form of AMD, the search for new therapeutic options continues to prevent the loss of photoreceptors and retinal pigment epithelium cells, characteristic of late stage AMD. In this review, we explain how mitochondrial dysfunction and endoplasmic reticulum stress participate in AMD pathogenesis. We also discuss a role of several antioxidants (bile acids, resveratrol, melatonin, humanin, and coenzyme Q10) in amelioration of AMD pathology.
Collapse
Affiliation(s)
- Valentina Bilbao-Malavé
- Department of Opthalmology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.B.-M.); (J.G.-Z.); (M.d.l.P.); (A.G.L.)
| | - Jorge González-Zamora
- Department of Opthalmology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.B.-M.); (J.G.-Z.); (M.d.l.P.); (A.G.L.)
| | - Miriam de la Puente
- Department of Opthalmology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.B.-M.); (J.G.-Z.); (M.d.l.P.); (A.G.L.)
| | - Sergio Recalde
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Universidad de Navarra, 31008 Pamplona, Spain; (S.R.); (P.F.-R.); (M.H.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain
- Red Temática de Investigación Cooperativa en Salud: ‘Prevention, Early Detection, and Treatment of the Prevalent Degenerative and Chronic Ocular Pathology’ from (RD16/0008/0011), Ministerio de Ciencia, Innovación y Universidades, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Patricia Fernandez-Robredo
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Universidad de Navarra, 31008 Pamplona, Spain; (S.R.); (P.F.-R.); (M.H.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain
- Red Temática de Investigación Cooperativa en Salud: ‘Prevention, Early Detection, and Treatment of the Prevalent Degenerative and Chronic Ocular Pathology’ from (RD16/0008/0011), Ministerio de Ciencia, Innovación y Universidades, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Hernandez
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Universidad de Navarra, 31008 Pamplona, Spain; (S.R.); (P.F.-R.); (M.H.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain
- Red Temática de Investigación Cooperativa en Salud: ‘Prevention, Early Detection, and Treatment of the Prevalent Degenerative and Chronic Ocular Pathology’ from (RD16/0008/0011), Ministerio de Ciencia, Innovación y Universidades, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alfredo Garcia Layana
- Department of Opthalmology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.B.-M.); (J.G.-Z.); (M.d.l.P.); (A.G.L.)
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Universidad de Navarra, 31008 Pamplona, Spain; (S.R.); (P.F.-R.); (M.H.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain
- Red Temática de Investigación Cooperativa en Salud: ‘Prevention, Early Detection, and Treatment of the Prevalent Degenerative and Chronic Ocular Pathology’ from (RD16/0008/0011), Ministerio de Ciencia, Innovación y Universidades, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Manuel Saenz de Viteri
- Department of Opthalmology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.B.-M.); (J.G.-Z.); (M.d.l.P.); (A.G.L.)
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Universidad de Navarra, 31008 Pamplona, Spain; (S.R.); (P.F.-R.); (M.H.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain
- Red Temática de Investigación Cooperativa en Salud: ‘Prevention, Early Detection, and Treatment of the Prevalent Degenerative and Chronic Ocular Pathology’ from (RD16/0008/0011), Ministerio de Ciencia, Innovación y Universidades, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
30
|
Costagliola G, Nuzzi G, Spada E, Comberiati P, Verduci E, Peroni DG. Nutraceuticals in Viral Infections: An Overview of the Immunomodulating Properties. Nutrients 2021; 13:nu13072410. [PMID: 34371920 PMCID: PMC8308811 DOI: 10.3390/nu13072410] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 12/21/2022] Open
Abstract
Nutraceuticals, including vitamin D, vitamin A, zinc, lactoferrin, polyphenols coenzyme Q, magnesium, and selenium, are implicated in the modulation of the complex molecular pathways involved in the immune response against viral pathogens. A common element of the activity of nutraceuticals is their ability to enhance the innate immune response against pathogens by acting on the major cellular subsets and inducing the release of pro-inflammatory cytokines and antimicrobial peptides. In some cases, this action is accompanied by a direct antimicrobial effect, as evidenced in the specific case of lactoferrin. Furthermore, nutraceuticals act through complex molecular mechanisms to minimize the damage caused by the activation of the immune system against pathogens, reducing the oxidative damage, influencing the antigen presentation, enhancing the differentiation and proliferation of regulatory T cells, driving the differentiation of lymphocyte subsets, and modulating the production of pro-inflammatory cytokines. In this paper, we review the main molecular mechanisms responsible for the immunomodulatory function of nutraceuticals, focusing on the most relevant aspects for the prevention and treatment of viral infections.
Collapse
Affiliation(s)
- Giorgio Costagliola
- Department of Clinical and Experimental Medicine, Division of Pediatrics, University of Pisa, Via Roma 57, 56126 Pisa, Italy; (G.C.); (G.N.); (E.S.); (P.C.)
| | - Giulia Nuzzi
- Department of Clinical and Experimental Medicine, Division of Pediatrics, University of Pisa, Via Roma 57, 56126 Pisa, Italy; (G.C.); (G.N.); (E.S.); (P.C.)
| | - Erika Spada
- Department of Clinical and Experimental Medicine, Division of Pediatrics, University of Pisa, Via Roma 57, 56126 Pisa, Italy; (G.C.); (G.N.); (E.S.); (P.C.)
| | - Pasquale Comberiati
- Department of Clinical and Experimental Medicine, Division of Pediatrics, University of Pisa, Via Roma 57, 56126 Pisa, Italy; (G.C.); (G.N.); (E.S.); (P.C.)
- Department of Clinical Immunology and Allergology, I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| | - Elvira Verduci
- Department of Pediatrics, San Paolo Hospital, 20142 Milan, Italy;
- Department of Health Science, University of Milan, 20142 Milan, Italy
| | - Diego G. Peroni
- Department of Clinical and Experimental Medicine, Division of Pediatrics, University of Pisa, Via Roma 57, 56126 Pisa, Italy; (G.C.); (G.N.); (E.S.); (P.C.)
- Correspondence: ; Tel.: +39-50-799-2100
| |
Collapse
|
31
|
Ubiquinol Supplementation of Donor Tissue Enhances Corneal Endothelial Cell Mitochondrial Respiration. Cornea 2021; 39:1285-1290. [PMID: 32558735 DOI: 10.1097/ico.0000000000002408] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE To determine whether ubiquinol improves mitochondrial function and cell viability in human donor corneal endothelial cells during hypothermic corneal tissue storage. METHODS Endothelial cell Descemet membrane tissues were treated with 10 μM ubiquinol, the reduced form of the antioxidant coenzyme Q10, for 5 days in Optisol-GS storage media before assaying for mitochondrial activity using extracellular flux analysis of oxygen consumption. In addition, endothelial cell Descemet membrane tissues were analyzed for cell viability using apoptosis and necrosis assays. Control tissues from mate corneas were treated with diluent only, and comparisons were analyzed for differences. RESULTS A total of 13 donor corneal tissues with a mean (SEM) preservation time of 11.8 days (0.4) were included for the analysis. Treatment with 10 μM ubiquinol increased spare respiratory capacity by 174% (P = 0.001), maximal respiration by 93% (P = 0.003), and proton leak by 80% (P = 0.047) compared with controls. Cells treated with ubiquinol had no significant change in cell necrosis or apoptosis. CONCLUSIONS Preliminary testing in donor corneal tissue at specified doses indicates that ubiquinol may be a useful biocompatible additive to hypothermic corneal storage media that increases corneal endothelial cell mitochondrial function. Additional investigations are indicated to further study and optimize the dose and formulation of ubiquinol for use in preserving donor corneal tissue function during hypothermic storage.
Collapse
|
32
|
Fernández-Del-Río L, Rodríguez-López S, Gutiérrez-Casado E, González-Reyes JA, Clarke CF, Burón MI, Villalba JM. Regulation of hepatic coenzyme Q biosynthesis by dietary omega-3 polyunsaturated fatty acids. Redox Biol 2021; 46:102061. [PMID: 34246922 PMCID: PMC8274332 DOI: 10.1016/j.redox.2021.102061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/29/2021] [Indexed: 11/24/2022] Open
Abstract
Dietary fats are important for human health, yet it is not fully understood how different fats affect various health problems. Although polyunsaturated fatty acids (PUFAs) are generally considered as highly oxidizable, those of the n-3 series can ameliorate the risk of many age-related disorders. Coenzyme Q (CoQ) is both an essential component of the mitochondrial electron transport chain and the only lipid-soluble antioxidant that animal cells can synthesize. Previous work has documented the protective antioxidant properties of CoQ against the autoxidation products of PUFAs. Here, we have explored in vitro and in vivo models to better understand the regulation of CoQ biosynthesis by dietary fats. In mouse liver, PUFAs increased CoQ content, and PUFAs of the n-3 series increased preferentially CoQ10. This response was recapitulated in hepatic cells cultured in the presence of lipid emulsions, where we additionally demonstrated a role for n-3 PUFAs as regulators of CoQ biosynthesis via the upregulation of several COQ proteins and farnesyl pyrophosphate levels. In both models, n-3 PUFAs altered the mitochondrial network without changing the overall mitochondrial mass. Furthermore, in cellular systems, n-3 PUFAs favored the synthesis of CoQ10 over CoQ9, thus altering the ratio between CoQ isoforms through a mechanism that involved downregulation of farnesyl diphosphate synthase activity. This effect was recapitulated by both siRNA silencing and by pharmacological inhibition of farnesyl diphosphate synthase with zoledronic acid. We highlight here the ability of n-3 PUFAs to regulate CoQ biosynthesis, CoQ content, and the ratio between its isoforms, which might be relevant to better understand the health benefits associated with this type of fat. Additionally, we identify for the first time zoledronic acid as a drug that inhibits CoQ biosynthesis, which must be also considered with respect to its biological effects on patients.
Collapse
Affiliation(s)
- Lucía Fernández-Del-Río
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario CeiA3, Córdoba, Spain; Department of Chemistry & Biochemistry, and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Sandra Rodríguez-López
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario CeiA3, Córdoba, Spain
| | - Elena Gutiérrez-Casado
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario CeiA3, Córdoba, Spain
| | - José Antonio González-Reyes
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario CeiA3, Córdoba, Spain
| | - Catherine F Clarke
- Department of Chemistry & Biochemistry, and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - María Isabel Burón
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario CeiA3, Córdoba, Spain
| | - José Manuel Villalba
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario CeiA3, Córdoba, Spain.
| |
Collapse
|
33
|
Navas P, Cascajo MV, Alcázar-Fabra M, Hernández-Camacho JD, Sánchez-Cuesta A, Rodríguez ABC, Ballesteros-Simarro M, Arroyo-Luque A, Rodríguez-Aguilera JC, Fernández-Ayala DJM, Brea-Calvo G, López-Lluch G, Santos-Ocaña C. Secondary CoQ 10 deficiency, bioenergetics unbalance in disease and aging. Biofactors 2021; 47:551-569. [PMID: 33878238 DOI: 10.1002/biof.1733] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/24/2021] [Indexed: 12/21/2022]
Abstract
Coenzyme Q10 (CoQ10 ) deficiency is a rare disease characterized by a decreased accumulation of CoQ10 in cell membranes. Considering that CoQ10 synthesis and most of its functions are carried out in mitochondria, CoQ10 deficiency cases are usually considered a mitochondrial disease. A relevant feature of CoQ10 deficiency is that it is the only mitochondrial disease with a successful therapy available, the CoQ10 supplementation. Defects in components of the synthesis machinery caused by mutations in COQ genes generate the primary deficiency of CoQ10 . Mutations in genes that are not directly related to the synthesis machinery cause secondary deficiency. Cases of CoQ10 deficiency without genetic origin are also considered a secondary deficiency. Both types of deficiency can lead to similar clinical manifestations, but the knowledge about primary deficiency is deeper than secondary. However, secondary deficiency cases may be underestimated since many of their clinical manifestations are shared with other pathologies. This review shows the current state of secondary CoQ10 deficiency, which could be even more relevant than primary deficiency for clinical activity. The analysis covers the fundamental features of CoQ10 deficiency, which are necessary to understand the biological and clinical differences between primary and secondary CoQ10 deficiencies. Further, a more in-depth analysis of CoQ10 secondary deficiency was undertaken to consider its origins, introduce a new way of classification, and include aging as a form of secondary deficiency.
Collapse
Affiliation(s)
- Plácido Navas
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - María V Cascajo
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - María Alcázar-Fabra
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan D Hernández-Camacho
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Sánchez-Cuesta
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Belén Cortés Rodríguez
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
- Laboratorio de Fisiopatología Celular y Bioenergética, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
| | - Manuel Ballesteros-Simarro
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Arroyo-Luque
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Carlos Rodríguez-Aguilera
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
- Laboratorio de Fisiopatología Celular y Bioenergética, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
| | - Daniel J M Fernández-Ayala
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Gloria Brea-Calvo
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Guillermo López-Lluch
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Santos-Ocaña
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
34
|
Rizzardi N, Liparulo I, Antonelli G, Orsini F, Riva A, Bergamini C, Fato R. Coenzyme Q10 Phytosome Formulation Improves CoQ10 Bioavailability and Mitochondrial Functionality in Cultured Cells. Antioxidants (Basel) 2021; 10:antiox10060927. [PMID: 34200321 PMCID: PMC8226950 DOI: 10.3390/antiox10060927] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/26/2021] [Accepted: 06/04/2021] [Indexed: 12/17/2022] Open
Abstract
Coenzyme Q10 (CoQ10) is a lipid-soluble molecule with a dual role: it transfers electrons in the mitochondrial transport chain by promoting the transmembrane potential exploited by the ATPase to synthesize ATP and, in its reduced form, is a membrane antioxidant. Since the high CoQ10 hydrophobicity hinders its bioavailability, several formulations have been developed to facilitate its cellular uptake. In this work, we studied the bioenergetic and antioxidant effects in I407 and H9c2 cells of a CoQ10 phytosome formulation (UBIQSOME®, UBQ). We investigated the cellular and mitochondrial content of CoQ10 and its redox state after incubation with UBQ. We studied different bioenergetic parameters, such as oxygen consumption, ATP content and mitochondrial potential. Moreover, we evaluated the effects of CoQ10 incubation on oxidative stress, membrane lipid peroxidation and ferroptosis and highlighted the connection between the intracellular concentration of CoQ10 and its antioxidant potency. Finally, we focused on the cellular mechanism that regulates UBQ internalization. We showed that the cell lines used in this work share the same uptake mechanism for UBQ, although the intestinal cell line was less efficient. Given the limitations of an in vitro model, the latter result supports that intestinal absorption is a critical step for the oral administration of Coenzyme Q10 formulations.
Collapse
Affiliation(s)
- Nicola Rizzardi
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, 6, 40126 Bologna, Italy; (N.R.); (I.L.); (G.A.); (R.F.)
| | - Irene Liparulo
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, 6, 40126 Bologna, Italy; (N.R.); (I.L.); (G.A.); (R.F.)
| | - Giorgia Antonelli
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, 6, 40126 Bologna, Italy; (N.R.); (I.L.); (G.A.); (R.F.)
| | | | - Antonella Riva
- Indena SpA, Viale Ortles, 20139 Milan, Italy; (F.O.); (A.R.)
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, 6, 40126 Bologna, Italy; (N.R.); (I.L.); (G.A.); (R.F.)
- Correspondence: ; Tel.: +39-051-209-1240
| | - Romana Fato
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, 6, 40126 Bologna, Italy; (N.R.); (I.L.); (G.A.); (R.F.)
| |
Collapse
|
35
|
Hidalgo-Gutiérrez A, González-García P, Díaz-Casado ME, Barriocanal-Casado E, López-Herrador S, Quinzii CM, López LC. Metabolic Targets of Coenzyme Q10 in Mitochondria. Antioxidants (Basel) 2021; 10:520. [PMID: 33810539 PMCID: PMC8066821 DOI: 10.3390/antiox10040520] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/14/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Coenzyme Q10 (CoQ10) is classically viewed as an important endogenous antioxidant and key component of the mitochondrial respiratory chain. For this second function, CoQ molecules seem to be dynamically segmented in a pool attached and engulfed by the super-complexes I + III, and a free pool available for complex II or any other mitochondrial enzyme that uses CoQ as a cofactor. This CoQ-free pool is, therefore, used by enzymes that link the mitochondrial respiratory chain to other pathways, such as the pyrimidine de novo biosynthesis, fatty acid β-oxidation and amino acid catabolism, glycine metabolism, proline, glyoxylate and arginine metabolism, and sulfide oxidation metabolism. Some of these mitochondrial pathways are also connected to metabolic pathways in other compartments of the cell and, consequently, CoQ could indirectly modulate metabolic pathways located outside the mitochondria. Thus, we review the most relevant findings in all these metabolic functions of CoQ and their relations with the pathomechanisms of some metabolic diseases, highlighting some future perspectives and potential therapeutic implications.
Collapse
Affiliation(s)
- Agustín Hidalgo-Gutiérrez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Pilar González-García
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - María Elena Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Eliana Barriocanal-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Sergio López-Herrador
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Catarina M. Quinzii
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Luis C. López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| |
Collapse
|
36
|
Stability of Reduced and Oxidized Coenzyme Q10 in Finished Products. Antioxidants (Basel) 2021; 10:antiox10030360. [PMID: 33673604 PMCID: PMC7997171 DOI: 10.3390/antiox10030360] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
The efficiency of coenzyme Q10 (CoQ10) supplements is closely associated with its content and stability in finished products. This study aimed to provide evidence-based information on the quality and stability of CoQ10 in dietary supplements and medicines. Therefore, ubiquinol, ubiquinone, and total CoQ10 contents were determined by a validated HPLC-UV method in 11 commercial products with defined or undefined CoQ10 form. Both forms were detected in almost all tested products, resulting in a total of CoQ10 content between 82% and 166% of the declared. Ubiquinol, ubiquinone, and total CoQ10 stability in these products were evaluated within three months of accelerated stability testing. Ubiquinol, which is recognized as the less stable form, was properly stabilized. Contrarily, ubiquinone degradation and/or reduction were observed during storage in almost all tested products. These reactions were also detected at ambient temperature within the products’ shelf-lives and confirmed in ubiquinone standard solutions. Ubiquinol, generated by ubiquinone reduction with vitamin C during soft-shell capsules’ storage, may lead to higher bioavailability and health outcomes. However, such conversion and inappropriate content in products, which specify ubiquinone, are unacceptable in terms of regulation. Therefore, proper CoQ10 stabilization through final formulations regardless of the used CoQ10 form is needed.
Collapse
|
37
|
López-Lluch G. Coenzyme Q homeostasis in aging: Response to non-genetic interventions. Free Radic Biol Med 2021; 164:285-302. [PMID: 33454314 DOI: 10.1016/j.freeradbiomed.2021.01.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/30/2020] [Accepted: 01/11/2021] [Indexed: 12/28/2022]
Abstract
Coenzyme Q (CoQ) is a key component for many essential metabolic and antioxidant activities in cells in mitochondria and cell membranes. Mitochondrial dysfunction is one of the hallmarks of aging and age-related diseases. Deprivation of CoQ during aging can be the cause or the consequence of this mitochondrial dysfunction. In any case, it seems clear that aging-associated CoQ deprivation accelerates mitochondrial dysfunction in these diseases. Non-genetic prolongevity interventions, including CoQ dietary supplementation, can increase CoQ levels in mitochondria and cell membranes improving mitochondrial activity and delaying cell and tissue deterioration by oxidative damage. In this review, we discuss the importance of CoQ deprivation in aging and age-related diseases and the effect of prolongevity interventions on CoQ levels and synthesis and CoQ-dependent antioxidant activities.
Collapse
Affiliation(s)
- Guillermo López-Lluch
- Universidad Pablo de Olavide, Centro Andaluz de Biología Del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Carretera de Utrera Km. 1, 41013, Sevilla, Spain.
| |
Collapse
|
38
|
Abstract
COPD represents a major cause of mortality and morbidity worldwide, is linked to systemic inflammation and tends to coexist with a variety of comorbidities. Inflammation, oxidative stress and protease-antiprotease imbalance represent the pathogenic triad of COPD. Even though oxidative stress and mitochondrial dysfunction is a well-studied phenomenon in COPD and there is a variety of studies that aim to counteract its effect, there is limited data available on the use of coenzyme Q10 in COPD. The aim of the current review is to analyze the current data on the use of coenzyme Q10 in the management of COPD and frequently encountered comorbidities.
Collapse
Affiliation(s)
- V I Zozina
- Department of Clinical Pharmacology and Propaedeutics of Internal Diseases, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - S Covantev
- Russian Medical Academy of Continuous Professional Education, Moscow, Russian Federation
| | - V G Kukes
- Department of Clinical Pharmacology and Propaedeutics of Internal Diseases, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - A Corlateanu
- Department of Respiratory Medicine, State University of Medicine and Pharmacy N. Testemitanu, Chisinau, Republic of Moldova
| |
Collapse
|
39
|
Castejón-Vega B, Battino M, Quiles JL, Bullon B, Cordero MD, Bullón P. Potential Role of the Mitochondria for the Dermatological Treatment of Papillon-Lefèvre. Antioxidants (Basel) 2021; 10:antiox10010095. [PMID: 33445524 PMCID: PMC7827181 DOI: 10.3390/antiox10010095] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 01/10/2023] Open
Abstract
The Papillon–Lefèvre syndrome (PLS) is a rare autosomal recessive disorder caused by mutations in the Cathepsin C (CTSC) gene, characterized by periodontitis and palmoplantar hyperkeratosis. The main inflammatory deficiencies include oxidative stress and autophagic dysfunction. Mitochondria are the main source of reactive oxygen species; their impaired function is related to skin diseases and periodontitis. The mitochondrial function has been evaluated in PLS and mitochondria have been targeted as a possible treatment for PLS. We show for the first time an important mitochondrial dysfunction associated with increased oxidative damage of mtDNA, reduced CoQ10 and mitochondrial mass and aberrant morphologies of the mitochondria in PLS patients. Mitochondrial dysfunction, determined by oxygen consumption rate (OCR) in PLS fibroblasts, was treated with CoQ10 supplementation, which determined an improvement in OCR and a remission of skin damage in a patient receiving a topical administration of a cream enriched with CoQ10 0.1%. We provide the first evidence of the role of mitochondrial dysfunction and CoQ10 deficiency in the pathophysiology of PLS and a future therapeutic option for PLS.
Collapse
Affiliation(s)
| | - Maurizio Battino
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Sez. Biochimica, Università Politecnica delle Marche, 60131 Ancona, Italy;
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - José L. Quiles
- Department of Physiology, Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center (CIBM), University of Granada, Avda. del Conocimiento s.n., 18100 Armilla, Spain;
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain
| | - Beatriz Bullon
- Department of Periodontology, Dental School, University of Sevilla, 41009 Sevilla, Spain;
| | - Mario D. Cordero
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, 11009 Cadiz, Spain
- Cátedra de Reproducción y Genética Humana del Instituto para el Estudio de la Biología de la Reproducción Humana (INEBIR)-Universidad Europea del Atlántico (UNEATLANTICO)-Fundación Universitaria Iberoamericana (FUNIBER), 39011 Santander, Spain
- Correspondence: (M.D.C.); (P.B.); Tel.: +34-9-5448-6783 (M.D.C.); +34-9-5448-1120 (P.B.)
| | - Pedro Bullón
- Department of Periodontology, Dental School, University of Sevilla, 41009 Sevilla, Spain;
- Correspondence: (M.D.C.); (P.B.); Tel.: +34-9-5448-6783 (M.D.C.); +34-9-5448-1120 (P.B.)
| |
Collapse
|
40
|
Webb M, Sideris DP. Intimate Relations-Mitochondria and Ageing. Int J Mol Sci 2020; 21:ijms21207580. [PMID: 33066461 PMCID: PMC7589147 DOI: 10.3390/ijms21207580] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction is associated with ageing, but the detailed causal relationship between the two is still unclear. We review the major phenomenological manifestations of mitochondrial age-related dysfunction including biochemical, regulatory and energetic features. We conclude that the complexity of these processes and their inter-relationships are still not fully understood and at this point it seems unlikely that a single linear cause and effect relationship between any specific aspect of mitochondrial biology and ageing can be established in either direction.
Collapse
Affiliation(s)
- Michael Webb
- Mitobridge Inc., an Astellas Company, 1030 Massachusetts Ave, Cambridge, MA 02138, USA
| | - Dionisia P Sideris
- Mitobridge Inc., an Astellas Company, 1030 Massachusetts Ave, Cambridge, MA 02138, USA
| |
Collapse
|
41
|
Wang Y, Xiao F, Jin G. Structural basis of quinolone derivatives, inhibition of type I and II topoisomerases and inquiry into the relevance of bioactivity in odd or even branches with molecular docking study. J Mol Struct 2020; 1221:128869. [PMID: 32834112 PMCID: PMC7347330 DOI: 10.1016/j.molstruc.2020.128869] [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: 05/18/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022]
Abstract
The structural modification of quinolone derivatives has been a hot spot in recent years, especially the modification of the N-1 position, which is the part that this article focuses on. In this paper, series of synthesized quinoline quaternary ammonium salts with odd and even carbon number alkyl groups in N-1 position were used to explain the influence of the alkyl side chain on activity. With respect to all the recently synthesized twenty products, the biological activity results exhibited significant antitumor and antibacterial activity with obvious differences in the target alkyliodine substituted compounds and the antibacterial activities apparently had the prominent odd-carbon number predominance. Compound 8-((4-(benzyloxy)phenyl)amino)-7-(ethoxycarbonyl)-5-propyl-[1,3]dioxolo[4,5-g]quinolin-5-ium (4d) was found to be the most potent derivative with IC50 values of 4 ± 0.88, 4 ± 0.42, 14±1.96, and 32±3.66 against A-549, Hela, SGC-7901, and L-02 cells, respectively, stronger than the positive control 5-FU and MTX. Furthermore, it had the most potent bacterial inhibitory activity of MIC value against E. coli (ATCC 29213) and Staphylococcus aureus (ATCC 8739) at 3.125 nmol mL−1. With respect to molecular simulations, in order to illustrate the possible mechanism of the difference between the series of compounds in the even or odd carbon chain alkyliodine substitution, this paper simulated the conceivable mode and explained the main interactions. Finally, we could find that the position and proportion of hydrogen bonds and other interactions in each series were regarded as the main reasons for this difference in activity.
Collapse
Affiliation(s)
- Yilin Wang
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, 212300, PR China
| | - Fuyan Xiao
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Guofan Jin
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| |
Collapse
|
42
|
Losson H, Gajulapalli SR, Lernoux M, Lee JY, Mazumder A, Gérard D, Seidel C, Hahn H, Christov C, Dicato M, Kirsch G, Han BW, Schnekenburger M, Diederich M. The HDAC6 inhibitor 7b induces BCR-ABL ubiquitination and downregulation and synergizes with imatinib to trigger apoptosis in chronic myeloid leukemia. Pharmacol Res 2020; 160:105058. [PMID: 32619722 DOI: 10.1016/j.phrs.2020.105058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 06/24/2020] [Accepted: 06/28/2020] [Indexed: 12/12/2022]
Abstract
Despite the discovery of tyrosine kinase inhibitors (TKIs) for the treatment of breakpoint cluster region-Abelson (BCR-ABL)+ cancer types, patients with chronic myeloid leukemia (CML) treated with TKIs develop resistance and severe adverse effects. Combination treatment, especially with a histone deacetylase (HDAC) 6 inhibitor (HDAC6i), appears to be an attractive option to prevent TKI resistance, considering the potential capacity of an HDAC6i to diminish BCR-ABL expression. We first validated the in vivo anti-cancer potential of the compound 7b by significantly reducing the tumor burden of BALB/c mice xenografted with K-562 cells, without notable organ toxicity. Here, we hypothesize that the HDAC6i compound 7b can lead to BCR-ABL downregulation in CML cells and sensitize them to TKI treatment. The results showed that combination treatment with imatinib and 7b resulted in strong synergistic caspase-dependent apoptotic cell death and drastically reduced the proportion of leukemia stem cells, whereas this treatment only moderately affected healthy cells. Ultimately, the combination significantly decreased colony formation in a semisolid methylcellulose medium and tumor mass in xenografted zebrafish compared to each compound alone. Mechanistically, the combination induced BCR-ABL ubiquitination and downregulation followed by disturbance of key proteins in downstream pathways involved in CML proliferation and survival. Taken together, our results suggest that an HDAC6i potentiates the effect of imatinib and could overcome TKI resistance in CML cells.
Collapse
Affiliation(s)
- Hélène Losson
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540, Luxembourg, Luxembourg
| | - Sruthi Reddy Gajulapalli
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Manon Lernoux
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540, Luxembourg, Luxembourg
| | - Jin-Young Lee
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Aloran Mazumder
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Déborah Gérard
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540, Luxembourg, Luxembourg
| | - Carole Seidel
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540, Luxembourg, Luxembourg
| | - Hyunggu Hahn
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Christo Christov
- Service d'Histologie, Faculté de Médicine, Université de Lorraine, INSERM U1256 NGERE, 54000, Nancy, France
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540, Luxembourg, Luxembourg
| | - Gilbert Kirsch
- UMR CNRS 7053 LC2M, Université de Lorraine, 57070, Metz, France
| | - Byung Woo Han
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Michael Schnekenburger
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540, Luxembourg, Luxembourg
| | - Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.
| |
Collapse
|
43
|
Fernández-del-Río L, Soubeyrand E, Basset GJ, Clarke CF. Metabolism of the Flavonol Kaempferol in Kidney Cells Liberates the B-ring to Enter Coenzyme Q Biosynthesis. Molecules 2020; 25:molecules25132955. [PMID: 32605010 PMCID: PMC7412559 DOI: 10.3390/molecules25132955] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/08/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
Coenzyme Q (CoQ) is an essential component of the mitochondrial electron transport chain and an important antioxidant present in all cellular membranes. CoQ deficiencies are frequent in aging and in age-related diseases, and current treatments are limited to CoQ supplementation. Strategies that rely on CoQ supplementation suffer from poor uptake and trafficking of this very hydrophobic molecule. In a previous study, the dietary flavonol kaempferol was reported to serve as a CoQ ring precursor and to increase the CoQ content in kidney cells, but neither the part of the molecule entering CoQ biosynthesis nor the mechanism were described. In this study, kaempferol labeled specifically in the B-ring was isolated from Arabidopsis plants. Kidney cells treated with this compound incorporated the B-ring of kaempferol into newly synthesized CoQ, suggesting that the B-ring is metabolized via a mechanism described in plant cells. Kaempferol is a natural flavonoid present in fruits and vegetables and possesses antioxidant, anticancer, and anti-inflammatory therapeutic properties. A better understanding of the role of kaempferol as a CoQ ring precursor makes this bioactive compound a potential candidate for the design of interventions aiming to increase endogenous CoQ biosynthesis and may improve CoQ deficient phenotypes in aging and disease.
Collapse
Affiliation(s)
- Lucía Fernández-del-Río
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
- Correspondence: (L.F.-d.-R.); (C.F.C.); Tel.: +1-(310)-825-0771 (C.F.C.)
| | - Eric Soubeyrand
- Department of Horticultural Sciences, University of Florida, Gainesville, FL 32611, USA; (E.S.); (G.J.B.)
| | - Gilles J. Basset
- Department of Horticultural Sciences, University of Florida, Gainesville, FL 32611, USA; (E.S.); (G.J.B.)
| | - Catherine F. Clarke
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
- Correspondence: (L.F.-d.-R.); (C.F.C.); Tel.: +1-(310)-825-0771 (C.F.C.)
| |
Collapse
|
44
|
Berardo A, Quinzii CM. Redefining infantile-onset multisystem phenotypes of coenzyme Q 10-deficiency in the next-generation sequencing era. JOURNAL OF TRANSLATIONAL GENETICS AND GENOMICS 2020; 4:22-35. [PMID: 33426503 PMCID: PMC7791541 DOI: 10.20517/jtgg.2020.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Primary coenzyme Q10 (CoQ10) deficiency encompasses a subset of mitochondrial diseases caused by mutations affecting proteins involved in the CoQ10 biosynthetic pathway. One of the most frequent clinical syndromes associated with primary CoQ10 deficiency is the severe infantile multisystemic form, which, until recently, was underdiagnosed. In the last few years, the availability of genetic screening through whole exome sequencing and whole genome sequencing has enabled molecular diagnosis in a growing number of patients with this syndrome and has revealed new disease phenotypes and molecular defects in CoQ10 biosynthetic pathway genes. Early genetic screening can rapidly and non-invasively diagnose primary CoQ10 deficiencies. Early diagnosis is particularly important in cases of CoQ10 deficient steroid-resistant nephrotic syndrome, which frequently improves with treatment. In contrast, the infantile multisystemic forms of CoQ10 deficiency, particularly when manifesting with encephalopathy, present therapeutic challenges, due to poor responses to CoQ10 supplementation. Administration of CoQ10 biosynthetic intermediate compounds is a promising alternative to CoQ10; however, further pre-clinical studies are needed to establish their safety and efficacy, as well as to elucidate the mechanism of actions of the intermediates. Here, we review the molecular defects causes of the multisystemic infantile phenotype of primary CoQ10 deficiency, genotype-phenotype correlations, and recent therapeutic advances.
Collapse
Affiliation(s)
- Andres Berardo
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Catarina M Quinzii
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
45
|
Comparative Bioavailability of Different Coenzyme Q10 Formulations in Healthy Elderly Individuals. Nutrients 2020; 12:nu12030784. [PMID: 32188111 PMCID: PMC7146408 DOI: 10.3390/nu12030784] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 12/19/2022] Open
Abstract
Coenzyme Q10 (CoQ10) plays a central role in mitochondrial oxidative phosphorylation. Several studies have shown the beneficial effects of dietary CoQ10 supplementation, particularly in relation to cardiovascular health. CoQ10 biosynthesis decreases in the elderly, and consequently, the beneficial effects of dietary supplementation in this population are of greater significance. However, most pharmacokinetic studies have been conducted on younger populations. The aim of this study was to investigate the single-dose bioavailability of different formulations of CoQ10 in a healthy geriatric population. A randomized, three-period, crossover bioavailability study was conducted on 21 healthy older adults (aged 65–74). The treatment was a single dose with a one-week washout period. Three different formulations containing the equivalent of 100 mg of CoQ10 were used: Q10Vital® water-soluble CoQ10 syrup (the investigational product—IP); ubiquinol capsules (the comparative product—CP); and ubiquinone capsules (the standard product—SP). Ubiquinone/ubiquinol was followed in the plasma for 48 h. An analysis of the ratio of the area under the baseline-corrected concentration curve (ΔAUC48) for total CoQ10 and a comparison to SP yielded the following: The bioavailability of CoQ10 in the IP was 2.4-fold higher (95% CI: 1.3–4.5; p = 0.002), while the bioavailability of ubiquinol (CP) was not significantly increased (1.7-fold; 95% CI: 0.9–3.1, p = 0.129). No differences in the redox status of the absorbed coenzyme Q10 were observed between formulations, showing that CoQ10 appeared in the blood mostly as ubiquinol, even if consumed as ubiquinone.
Collapse
|
46
|
Pergolizzi JV, Coluzzi F, Colucci RD, Olsson H, LeQuang JA, Al-Saadi J, Magnusson P. Statins and muscle pain. Expert Rev Clin Pharmacol 2020; 13:299-310. [PMID: 32089020 DOI: 10.1080/17512433.2020.1734451] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Statins remain among the most frequently prescribed drugs and constitute a cornerstone in the prevention of cardiovascular disease. However, muscle symptoms are often reported from patients on statins. Muscle symptoms are frequently reported as adverse events associated with statin therapy.Areas covered: In the present narrative review, statin-associated muscle pain is discussed. It elucidates potential mechanisms and possible targets for management.Expert opinion: In general, the evidence in support of muscle pain caused by statins is in some cases equivocal and not particularly strong. Reported symptoms are difficult to quantify. Rarely is it possible to establish a causal link between statins and muscle pain. In randomized controlled trials, statins are well tolerated, and muscle-pain related side-effects is similar to placebo. There are also nocebo effects of statins. Exchange of statin may be beneficial although all statins have been associated with muscle pain. In some patients reduction of dose is worth trying, especially in primary prevention Although the benefits of statins outweigh potential risks in the vast majority of cases, careful clinical judgment may be necessary in certain cases to manage potential side effects on an individual basis.
Collapse
Affiliation(s)
| | - Flaminia Coluzzi
- Department of Medical and Surgical Sciences and Biotechnologies, Unit of Anaesthesia, Intensive Care and Pain Medicine, Sapienza University of Rome, Rome, Italy
| | - Robert D Colucci
- NEMA Research, Inc., Naples, FL, USA.,Colucci & Associates, LLC, Newtown, Connecticut, USA
| | - Hanna Olsson
- Centre for Research and Development, Region Gävleborg/Uppsala University, Gävle, Sweden
| | | | - Jonathan Al-Saadi
- Centre for Research and Development, Region Gävleborg/Uppsala University, Gävle, Sweden
| | - Peter Magnusson
- Centre for Research and Development, Region Gävleborg/Uppsala University, Gävle, Sweden.,Cardiology Research Unit, Institution of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|