1
|
Moiseeva NV, Sokolov AE, Trushkov IV, Kokorekin VA. Electrochemically driven Michael reaction: synthesis of hydroquinone thioethers. Org Biomol Chem 2025; 23:1089-1093. [PMID: 39692761 DOI: 10.1039/d4ob01886a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
An original method for the synthesis of a diverse array of hydroquinone thioethers with yields of 36% to 99% under mild conditions is described. The process is voltammetrically controlled and involves electrogenerating active protonated p-quinone followed by thiol addition.
Collapse
Affiliation(s)
- Natalia V Moiseeva
- N. D. Zelinsky Institute of Organic Chemistry, Leninsky prosp. 47, Moscow 119991, Russian Federation.
| | - Alexey E Sokolov
- N. D. Zelinsky Institute of Organic Chemistry, Leninsky prosp. 47, Moscow 119991, Russian Federation.
| | - Igor V Trushkov
- N. D. Zelinsky Institute of Organic Chemistry, Leninsky prosp. 47, Moscow 119991, Russian Federation.
| | - Vladimir A Kokorekin
- N. D. Zelinsky Institute of Organic Chemistry, Leninsky prosp. 47, Moscow 119991, Russian Federation.
- Sechenov First Moscow State Medical University, Trubetskaya st. 8-2, Moscow 119991, Russian Federation
| |
Collapse
|
2
|
Mansour DF, Hashad IM, Rady M, Abd-El Razik AN, Saleh DO. Diosmin and Coenzyme q10: Synergistic histopathological and functional protection against doxorubicin-induced hepatorenal injury in rats. Toxicol Rep 2024; 13:101848. [PMID: 39703765 PMCID: PMC11655815 DOI: 10.1016/j.toxrep.2024.101848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/23/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024] Open
Abstract
Doxorubicin (DOX) is a cytotoxic anthracycline used to treat a variety of cancers. Cardiotoxicity, hepatotoxicity, and nephrotoxicity are adverse effects of DOX, that limit prognosis. The study aims to determine if diosmin (DIOS) and coenzyme Q10 (CoQ10) alone or in combination protect rats against DOX-induced liver and kidney damage. Adult male rats were assigned randomly in five groups. An intraperitoneal injection of DOX (2.5 mg/kg) was given to the DOX group every other day for three weeks, whereas a normal control group received the vehicle. Diosmin group received oral DIOS (100 mg/kg), Co-Q10 group received oral CoQ10 (10 mg/kg) and combination group received oral DIOS and CoQ10 daily for three weeks concomitantly with DOX. Sera and tissues were obtained 24 hours after last DOX injection. Serum aspartate transaminase (AST), alanine transaminase (ALT), creatinine, urea, total bilirubin and direct bilirubin were detected with hepatic and renal reduced glutathione (GSH), malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-α) and nuclear factor kappa-B (NF-κB). Histopathology and morphometry of liver and kidney were assessed. DOX exerted significant hepatorenal toxicity via elevation of liver and kidney functions, inducing oxidative stress by reducing GSH and elevating MDA, triggering renal and hepatic TNF-α and NF-kB. DIOS and CoQ10 modulated hepatic and renal functions, oxidative stress and inflammatory biomarkers. DIOS-CoQ10 combination treatment showed significant improvement in histopathology of liver and kidney along with morphometry compared to DOX group. In conclusion, combining DIOS and CoQ10 exhibited synergistic protective activity against DOX-induced hepatic and renal insult via their antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Dina F. Mansour
- Pharmacology Department, Medical Research and Clinical Studies Institute - National Research Centre, Dokki, Giza 12622, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Galala University, Mount Ataka, Suez, Egypt
| | - Ingy M. Hashad
- Department of Biochemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Mona Rady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, The German University in Cairo, Cairo, Egypt
- Faculty of Biotechnology, German International University, New Administrative Capital, Cairo, Egypt
| | - Amira N. Abd-El Razik
- Pathology Department, Medical Research and Clinical Studies Institute - National Research Centre, Dokki, Giza 12622, Egypt
| | - Dalia O. Saleh
- Pharmacology Department, Medical Research and Clinical Studies Institute - National Research Centre, Dokki, Giza 12622, Egypt
| |
Collapse
|
3
|
Maciejewska-Stupska K, Czarnecka K, Szymański P. Bioavailability enhancement of coenzyme Q 10: An update of novel approaches. Arch Pharm (Weinheim) 2024; 357:e2300676. [PMID: 38683827 DOI: 10.1002/ardp.202300676] [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: 11/19/2023] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 05/02/2024]
Abstract
Coenzyme Q10 (CoQ10) is an essential, lipid-soluble vitamin involved in electron transport in the oxidoreductive reactions of the mitochondrial respiratory chain. Structurally, the quinone ring is connected to an isoprenoid moiety, which has a high molecular weight. Over the years, coenzyme Q10 has become relevant in the treatment of several diseases, like neurodegenerative disorders, coronary diseases, diabetes, hypercholesterolemia, cancer, and others. According to studies, CoQ10 supplementation might be beneficial in the treatment of CoQ10 deficiencies and disorders associated with oxidative stress. However, the water-insoluble nature of CoQ10 is a major hindrance to successful supplementation. So far, many advancements in CoQ10 bioavailability enhancement have been developed using novel drug carriers such as solid dispersion, liposomes, micelles, nanoparticles, nanoemulsions, self-emulsifying drug systems, or various innovative approaches (CoQ10 complexation with proteins). This article aims to provide an update on methods to improve CoQ10 solubility and bioavailability.
Collapse
Affiliation(s)
- Karolina Maciejewska-Stupska
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Lodz, Poland
| | - Kamila Czarnecka
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Lodz, Poland
| | - Paweł Szymański
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Lodz, Poland
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| |
Collapse
|
4
|
Yuan S, Che Y, Wang Z, Xing K, Xie X, Chen Y. Mitochondrion-targeted carboxymethyl chitosan hybrid nanoparticles loaded with Coenzyme Q10 protect cardiac grafts against cold ischaemia‒reperfusion injury in heart transplantation. J Transl Med 2023; 21:925. [PMID: 38124174 PMCID: PMC10734076 DOI: 10.1186/s12967-023-04763-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Heart transplantation (HT) has been approved as an optimal therapeutic regimen for patients with terminal-stage cardiac failure. However, cold ischaemia‒reperfusion (I/R) injury remains an unavoidable and outstanding challenge, which is a major factor in early graft dysfunction and an obstacle to long-term survival in HT. Cold I/R injury induces cardiac graft injury by promoting mitochondrial dysfunction and augmenting free radical production and inflammatory responses. We therefore designed a mitochondrion-targeted nanocarrier loaded with Coenzyme Q10 (CoQ10) (CoQ10@TNPs) for treatment of cold I/R injury after cardiac graft in a murine heterotopic cardiac transplantation model. METHODS Hybrid nanoparticles composed of CaCO3/CaP/biotinylated-carboxymethylchitosan (CaCO3/CaP/BCMC) were synthesized using the coprecipitation method, and the mitochondria-targeting tetrapeptide SS31 was incorporated onto the surface of the hybrid nanoparticles through biotin-avidin interactions. Transmission electron microscopy (TEM) and dynamic light scattering (DLS) analysis were used for characterisation. In vitro, the hypoxia-reoxygenation model of H9c2 cells was employed to replicate in vivo cold I/R injury and treated with CoQ10@TNPs. The impact of CoQ10@TNPs on H9c2 cell injury was assessed by analysis of oxidative damage and apoptosis. In vivo, donor hearts (DHs) were perfused with preservation solution containing CoQ10@TNPs and stored in vitro at 4 °C for 12 h. The DHs were heterotopically transplanted and analysed for graft function, oxidative damage, apoptosis, and inflammatory markers 1 day post-transplantation. RESULTS CoQ10@TNPs were successfully synthesized and delivered CoQ10 to the mitochondria of the cold ischaemic myocardium. In vitro experiments demonstrated that CoQ10@TNPs was taken up by H9c2 cells at 4 °C and localized within the mitochondria, thus ameliorating oxidative stress damage and mitochondrial injury in cold I/R injury. In vivo experiments showed that CoQ10@TNPs accumulated in DH tissue at 4 °C, localized within the mitochondria during cold storage and improved cardiac graft function by attenuating mitochondrial oxidative injury and inflammation. CONCLUSIONS CoQ10@TNPs can precisely deliver CoQ10 to the mitochondria of cold I/R-injured cardiomyocytes to effectively eliminate mitochondrial reactive oxygen species (mtROS), thus reducing oxidative injury and inflammatory reactions in cold I/R-injured graft tissues and finally improving heart graft function. Thus, CoQ10@TNPs offer an effective approach for safeguarding cardiac grafts against extended periods of cold ischaemia, emphasizing the therapeutic potential in mitigating cold I/R injury during HT. These findings present an opportunity to enhance existing results following HT and broaden the range of viable grafts for transplantation.
Collapse
Affiliation(s)
- Shun Yuan
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanjia Che
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, Hubei, People's Republic of China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China.
| | - Kai Xing
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoping Xie
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanyang Chen
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Lee WE, Genetzakis E, Figtree GA. Novel Strategies in the Early Detection and Treatment of Endothelial Cell-Specific Mitochondrial Dysfunction in Coronary Artery Disease. Antioxidants (Basel) 2023; 12:1359. [PMID: 37507899 PMCID: PMC10376062 DOI: 10.3390/antiox12071359] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Although elevated cholesterol and other recognised cardiovascular risk factors are important in the development of coronary artery disease (CAD) and heart attack, the susceptibility of humans to this fatal process is distinct from other animals. Mitochondrial dysfunction of cells in the arterial wall, particularly the endothelium, has been strongly implicated in the pathogenesis of CAD. In this manuscript, we review the established evidence and mechanisms in detail and explore the potential opportunities arising from analysing mitochondrial function in patient-derived cells such as endothelial colony-forming cells easily cultured from venous blood. We discuss how emerging technology and knowledge may allow us to measure mitochondrial dysfunction as a potential biomarker for diagnosis and risk management. We also discuss the "pros and cons" of animal models of atherosclerosis, and how patient-derived cell models may provide opportunities to develop novel therapies relevant for humans. Finally, we review several targets that potentially alleviate mitochondrial dysfunction working both via direct and indirect mechanisms and evaluate the effect of several classes of compounds in the cardiovascular context.
Collapse
Affiliation(s)
- Weiqian E. Lee
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Elijah Genetzakis
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Gemma A. Figtree
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW 2065, Australia
| |
Collapse
|
6
|
Zhang T, He Q, Xiu H, Zhang Z, Liu Y, Chen Z, Hu H. Efficacy and Safety of Coenzyme Q10 Supplementation in the Treatment of Polycystic Ovary Syndrome: a Systematic Review and Meta-analysis. Reprod Sci 2023; 30:1033-1048. [PMID: 35941510 DOI: 10.1007/s43032-022-01038-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 07/12/2022] [Indexed: 11/27/2022]
Abstract
The aim of this study is to evaluate the efficacy and safety of coenzyme Q10 supplementation in the treatment of polycystic ovary syndrome (PCOS). We first searched PubMed, Wanfang Data, CNKI, Embase, ClinicalTrial.gov, and other databases. The retrieval time from the establishment of the database to January 2021. We collected relevant randomized controlled trials (RCTs) about coenzyme Q10 in the treatment of PCOS. Risk of bias assessment and meta-analysis of RCTs were performed using RevMan 5.0 software. This systematic review and meta-analysis include a total of 9 RCTs involving 1021 patients. The results show that the addition of coenzyme Q10 may improve insulin resistance (HOMA-IR (WMD - 0.67 [- 0.87, - 0.48], P < 0.00001); fasting insulin (WMD - 1.75 [- 2.65, - 0.84], P = 0.0002); fasting plasma glucose (WMD - 5.20 [- 8.86, - 1.54], P = 0.005)), improve sex hormone levels (FSH (SMD - 0.45 [0.11, 0.78], P = 0.009); testosterone (SMD - 0.28 [- 0.49, - 0.06], P = 0.01)), and improve blood lipids (triglycerides (SMD - 0.49 [- 0.89, - 0.09], P = 0.02); total cholesterol (SMD - 0.35 [- 0.56, - 0.14], P = 0.001); LDL-C (SMD - 0.22 [- 0.43, - 0.01], P = 0.04); HDL-C (SMD 0.22 [0.01, 0.43], P = 0.04)). Only one RCT reported adverse events, and they found that patients had no adverse effects or symptoms following supplementation. Based on the current evidence, it could be considered that the addition of CoQ10 is a safe therapy to improve PCOS by improving insulin resistance (reduce HOMA-IR, FINS, FPG), increasing sex hormone levels (increase FSH, reduce testosterone), and improving blood lipids (reduce TG, TC, LDL-C, and increased HDL-C).
Collapse
Affiliation(s)
- Tianqing Zhang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Qi He
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Hao Xiu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - ZiZhu Zhang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Yao Liu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Zhenrong Chen
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
| | - Hengjing Hu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China.
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
7
|
Mitochondrial Genetic Background May Impact Statins Side Effects and Atherosclerosis Development in Familial Hypercholesterolemia. Int J Mol Sci 2022; 24:ijms24010471. [PMID: 36613915 PMCID: PMC9820128 DOI: 10.3390/ijms24010471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/30/2022] Open
Abstract
Heredity of familial hypercholesterolemia (FH) can present as a dominant monogenic disorder of polygenic origin or with no known genetic cause. In addition, the variability of the symptoms among individuals or within the same families evidence the potential contribution of additional factors than monogenic mutations that could modulate the development and severity of the disease. In addition, statins, the lipid-lowering drugs which constitute the first-line therapy for the disease, cause associated muscular symptoms in a certain number of individuals. Here, we analyze the evidence of the mitochondrial genetic variation with a special emphasis on the role of CoQ10 to explain this variability found in both disease symptoms and statins side effects. We propose to use mtDNA variants and copy numbers as markers for the cardiovascular disease development of FH patients and to predict potential statin secondary effects and explore new mechanisms to identify new markers of disease or implement personalized medicine strategies for FH therapy.
Collapse
|
8
|
Tan M, Zhang HB, Ye PP, Cui FJ, Chen C, Zhou TL, Shi JC, Zhang W, Shu XQ, Chen ZW. Distinguishing strong, mellow and light fragrant rapeseed oils in China using physicochemical, nutritional and aroma profiles. JOURNAL OF FOOD MEASUREMENT AND CHARACTERIZATION 2022. [DOI: 10.1007/s11694-022-01729-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
9
|
Wu YL, Chang JC, Sun HL, Cheng WL, Yen YP, Lin YS, Chao YC, Liu KH, Huang CS, Liu KL, Liu CS. Coenzyme Q10 Supplementation Increases Removal of the ATXN3 Polyglutamine Repeat, Reducing Cerebellar Degeneration and Improving Motor Dysfunction in Murine Spinocerebellar Ataxia Type 3. Nutrients 2022; 14:nu14173593. [PMID: 36079853 PMCID: PMC9459709 DOI: 10.3390/nu14173593] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Coenzyme Q10 (CoQ10), a well-known antioxidant, has been explored as a treatment in several neurodegenerative diseases, but its utility in spinocerebellar ataxia type 3 (SCA3) has not been explored. Herein, the protective effect of CoQ10 was examined using a transgenic mouse model of SCA3 onset. These results demonstrated that a diet supplemented with CoQ10 significantly improved murine locomotion, revealed by rotarod and open-field tests, compared with untreated controls. Additionally, a histological analysis showed the stratification of cerebellar layers indistinguishable from that of wild-type littermates. The increased survival of Purkinje cells was reflected by the reduced abundance of TUNEL-positive nuclei and apoptosis markers of activated p53, as well as lower levels of cleaved caspase 3 and cleaved poly-ADP-ribose polymerase. CoQ10 effects were related to the facilitation of the autophagy-mediated clearance of mutant ataxin-3 protein, as evidenced by the increased expression of heat shock protein 27 and autophagic markers p62, Beclin-1 and LC3II. The expression of antioxidant enzymes heme oxygenase 1 (HO-1), glutathione peroxidase 1 (GPx1) and superoxide dismutase 1 (SOD1) and 2 (SOD2), but not of glutathione peroxidase 2 (GPx2), were restored in 84Q SCA3 mice treated with CoQ10 to levels even higher than those measured in wild-type control mice. Furthermore, CoQ10 treatment also prevented skeletal muscle weight loss and muscle atrophy in diseased mice, revealed by significantly increased muscle fiber area and upregulated muscle protein synthesis pathways. In summary, our results demonstrated biochemical and pharmacological bases for the possible use of CoQ10 in SCA3 therapy.
Collapse
Affiliation(s)
- Yu-Ling Wu
- Vascular and Genomic Center, Institute of ATP, Changhua Christian Hospital, Changhua 50091, Taiwan
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Jui-Chih Chang
- Center of Regenerative Medicine and Tissue Repair, Changhua Christian Hospital, Changhua 50091, Taiwan
- General Research Laboratory of Research Department, Changhua Christian Hospital, Changhua 50091, Taiwan
| | - Hai-Lun Sun
- School of Medicine, Chung Shan Medical University, Taichung 40203, Taiwan
- Department of Pediatrics, Division of Allergy, Asthma and Rheumatology, Chung Shan Medical University Hospital, Taichung 40203, Taiwan
| | - Wen-Ling Cheng
- Vascular and Genomic Center, Institute of ATP, Changhua Christian Hospital, Changhua 50091, Taiwan
| | - Yu-Pei Yen
- Department of Nutrition, Chung Shan Medical University, Taichung 40203, Taiwan
| | - Yong-Shiou Lin
- Vascular and Genomic Center, Institute of ATP, Changhua Christian Hospital, Changhua 50091, Taiwan
| | - Yi-Chun Chao
- Inflammation Research & Drug Development Center, Changhua Christian Hospital, Changhua 50091, Taiwan
| | - Ko-Hung Liu
- Inflammation Research & Drug Development Center, Changhua Christian Hospital, Changhua 50091, Taiwan
| | - Ching-Shan Huang
- Center of Regenerative Medicine and Tissue Repair, Changhua Christian Hospital, Changhua 50091, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung 40203, Taiwan
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung 40203, Taiwan
- Correspondence: (K.-L.L.); (C.-S.L.); Tel.: +886-4-24730022 (ext. 12136) (K.-L.L.); +886-4-7238595 (ext. 4751) (C.-S.L.)
| | - Chin-San Liu
- Vascular and Genomic Center, Institute of ATP, Changhua Christian Hospital, Changhua 50091, Taiwan
- Department of Neurology, Changhua Christian Hospital, Changhua 50094, Taiwan
- Graduate Institute of Integrated Medicine College of Chinese Medicine, China Medical University, Taichung 40447, Taiwan
- College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Correspondence: (K.-L.L.); (C.-S.L.); Tel.: +886-4-24730022 (ext. 12136) (K.-L.L.); +886-4-7238595 (ext. 4751) (C.-S.L.)
| |
Collapse
|
10
|
Stamerra CA, Di Giosia P, Giorgini P, Ferri C, Sukhorukov VN, Sahebkar A. Mitochondrial Dysfunction and Cardiovascular Disease: Pathophysiology and Emerging Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9530007. [PMID: 35958017 PMCID: PMC9363184 DOI: 10.1155/2022/9530007] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022]
Abstract
Mitochondria ensure the supply of cellular energy through the production of ATP via oxidative phosphorylation. The alteration of this process, called mitochondrial dysfunction, leads to a reduction in ATP and an increase in the production of reactive oxygen species (ROS). Mitochondrial dysfunction can be caused by mitochondrial/nuclear DNA mutations, or it can be secondary to pathological conditions such as cardiovascular disease, aging, and environmental stress. The use of therapies aimed at the prevention/correction of mitochondrial dysfunction, in the context of the specific treatment of cardiovascular diseases, is a topic of growing interest. In this context, the data are conflicting since preclinical studies are numerous, but there are no large randomized studies.
Collapse
Affiliation(s)
- Cosimo Andrea Stamerra
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
- Department of Internal Medicine, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Paolo Di Giosia
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
- Department of Internal Medicine, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Paolo Giorgini
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
| | - Claudio Ferri
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
| | - Vasily N. Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, Moscow 121609, Russia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Preclinical and Clinical Role of Coenzyme Q10 Supplementation in Various Pathological States. Drug Res (Stuttg) 2022; 72:367-371. [PMID: 35724675 DOI: 10.1055/a-1835-1738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Coenzyme Q10 (CoQ10) is an efficient antioxidant produced endogenously in a living organism. It acts as an important cofactor in the electron transport system of mitochondria and reported as a safe supplement in humans and animals with minimal adverse effect. CoQ10 is found naturally, as a trans configuration, chemical nomenclature of which is 2,3- dimethoxy-5- methyl-6-decaprenyle -1,4-benzoquinone. It is found in the body in two forms. In quinone form (oxidized form), it serves as an electron transporter that transfers the electrons in the electron transport chain between various complexes, and in ubiquinol form (reduced form), it serves as potent antioxidants by scavenging free radicals or by tocopherol regeneration in the living organism. Its primary roles include synthesis of adenosine triphosphate (ATP), stabilizes lipid membrane, antioxidant activity, cell growth stimulation, and cell death inhibition. CoQ10 has shown a variety of pharmacological and clinical effects including neuroprotective, hepatoprotective, anti-atherosclerotic, anticonvulsant, antidepressant, anti-inflammatory, antinociceptive, cardiovascular, antimicrobial, immunomodulatory, and various effects on the central nervous system. Present review has set about to bring updated information regarding to clinical and preclinical activities of CoQ10, which may be helpful to researchers to explore a new bioactive molecules for various therapeutic application.
Collapse
|
12
|
Tan M, Chen C, Fu X, Cui FJ, Zhang HB, Ye PP, Zhang W, Shu XQ, Shi JC, Chen ZW. Roasting Treatments Affect Physicochemical, Aroma and Nutritional Quality of Strong Fragrant Rapeseed Oil. J Food Compost Anal 2022. [DOI: 10.1016/j.jfca.2022.104648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
13
|
Combined drug triads for synergic neuroprotection in retinal degeneration. Biomed Pharmacother 2022; 149:112911. [DOI: 10.1016/j.biopha.2022.112911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/23/2022] Open
|
14
|
Liu Z, Li Y, Li C, Yu L, Chang Y, Qu M. Delivery of coenzyme Q10 with mitochondria-targeted nanocarrier attenuates renal ischemia-reperfusion injury in mice. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112536. [PMID: 34857313 DOI: 10.1016/j.msec.2021.112536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/18/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022]
Abstract
Ischemia-reperfusion (I/R) injury causes high morbidity, mortality, and healthcare costs. I/R induces acute kidney injury through exacerbating the mitochondrial damage and increasing inflammatory and oxidative responses. Here, we developed the mitochondria-targeted nanocarrier to delivery of Coenzyme Q10 (CoQ10) for renal I/R treatment in animal model. The mitochondria-targeted TPP CoQ10 nanoparticles (T-NPCoQ10) were synthesized through ABC miktoarm polymers method and characterized by dynamic light scattering (DLS) and transmission electron microscopy (TEM). The I/R mouse model and oxygen-glucose deprivation/reperfusion (D/R) model were created to examine the role of T-NPCoQ10 on renal I/R. Mitochondrial DNA damage, apoptosis, and inflammatory cytokines were measured in I/R injury mice. Plasma creatinine, urea nitrogen, tubular injury score was tested to assess the renal function. T-NPCoQ10 nanoparticles could be delivered to renal mitochondria preciously and efficiently. T-NPCoQ10 administration attenuated oxidative injury in both cell and animal models significantly, alleviated mtDNA damage, suppressed inflammatory and apoptotic responses, and improved renal function. The mitochondria specific CoQ10 delivery provided a precious and efficient method for protecting inflammatory and oxidative responses of I/R-induced renal damage.
Collapse
Affiliation(s)
- Zhaohui Liu
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China.
| | - Yan Li
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China
| | - Chunlei Li
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China
| | - Lili Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China
| | - Yulin Chang
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China
| | - Min Qu
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China
| |
Collapse
|
15
|
Yang HL, Tsai CH, Shrestha S, Lee CC, Liao JW, Hseu YC. Coenzyme Q 0, a novel quinone derivative of Antrodia camphorata, induces ROS-mediated cytotoxic autophagy and apoptosis against human glioblastoma cells in vitro and in vivo. Food Chem Toxicol 2021; 155:112384. [PMID: 34229024 DOI: 10.1016/j.fct.2021.112384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/29/2021] [Accepted: 07/02/2021] [Indexed: 11/22/2022]
Abstract
Coenzyme Q0 (CoQ0, 2,3-dimethoxy-5-methyl-1,4-benzoquinone) derived from Antrodia camphorata exerts anticancer activities against breast, melanoma, and ovarian carcinoma. Glioblastoma multiforme is a common tumor affecting the central nervous system. This study explored anticancer properties of CoQ0 on human glioblastoma both in vitro and in vivo, and explained the molecular mechanism behind it. CoQ0 treatment retarded the growth and suppressed colony formation in glioblastoma (U87MG and GBM8401) cells. CoQ0 induced apoptosis by activation of caspase-3, cleavage of PARP, and dysregulation of Bax and Bcl-2 in both cell lines. Annexin V/PI staining indicated CoQ0 mediated necrosis and apoptosis. Interestingly, AVOs were increased trough induction of autophagy by CoQ0, LC3-II accumulation, and p62/SQSTM1 expression, leading to death mechanism. Z-VAD-FMK has no effect on CoQ0-induced autophagy but autophagy inhibition by 3-methyladenine (3-MA)/chloroquine (CQ) led to CoQ0-induced apoptosis. N-acetylcysteine (NAC) inhibited CoQ0-mediated ROS production and diminished CoQ0-induced apoptotic and autophagic cell death. Further, CoQ0 inhibited PI3K/AKT/mTOR signaling pathways. CoQ0 reduced the tumor burden in U87MG and GBM8401 xenografted athymic nude mice and significantly modulated tumor xenograft by inducing apoptosis and autophagy. CoQ0 generated ROS-mediated apoptotic and autophagic cell death for effective glioblastoma treatment.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Institute of Nutrition, College of Health Care, China Medical University, Taichung, 40402, Taiwan
| | - Chia-Hsuan Tsai
- Institute of Nutrition, College of Health Care, China Medical University, Taichung, 40402, Taiwan
| | - Sirjana Shrestha
- Institute of Nutrition, College of Health Care, China Medical University, Taichung, 40402, Taiwan
| | - Chuan-Chen Lee
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, 41354, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung-Hsing University, Taichung, 402, Taiwan
| | - You-Cheng Hseu
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung, 40402, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, 41354, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan; Research Center of Chinese Herbal Medicine, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
16
|
Alam M, Yadav RK, Minj E, Tiwari A, Mehan S. Exploring Molecular Approaches in Amyotrophic Lateral Sclerosis: Drug Targets from Clinical and Pre-Clinical Findings. Curr Mol Pharmacol 2021; 14:263-280. [PMID: 32342825 DOI: 10.2174/1566524020666200427214356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/24/2019] [Accepted: 12/26/2019] [Indexed: 11/22/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease (MND) characterized by the death of upper and lower motor neurons (corticospinal tract) in the motor cortex, basal ganglia, brain stem, and spinal cord. The patient experiences the sign and symptoms between 55 to 75 years of age, which include impaired motor movement, difficulty in speaking and swallowing, grip loss, muscle atrophy, spasticity, and sometimes associated with memory and cognitive impairments. Median survival is 3 to 5 years after diagnosis and 5 to 10% of the patients live for more than 10 years. The limited intervention of pharmacologically active compounds, that are used clinically, is majorly associated with the narrow therapeutic index. Pre-clinically established experimental models, where neurotoxin methyl mercury mimics the ALS like behavioural and neurochemical alterations in rodents associated with neuronal mitochondrial dysfunctions and downregulation of adenyl cyclase mediated cAMP/CREB, is the main pathological hallmark for the progression of ALS in central as well in the peripheral nervous system. Despite the considerable investigation into neuroprotection, it still constrains treatment choices to strong care and organization of ALS complications. Therefore, this current review specially targeted the investigation of clinical and pre-clinical features available for ALS to understand the pathogenic mechanisms and to explore the pharmacological interventions associated with the up-regulation of intracellular adenyl cyclase/cAMP/ CREB and activation of mitochondrial-ETC coenzyme-Q10 as a future drug target in the amelioration of ALS mediated motor neuronal dysfunctions.
Collapse
Affiliation(s)
- Mamtaj Alam
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Rajeshwar K Yadav
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Elizabeth Minj
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Aarti Tiwari
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Sidharth Mehan
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| |
Collapse
|
17
|
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: 21] [Impact Index Per Article: 5.3] [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
|
18
|
Gutierrez-Mariscal FM, de la Cruz-Ares S, Torres-Peña JD, Alcalá-Diaz JF, Yubero-Serrano EM, López-Miranda J. Coenzyme Q 10 and Cardiovascular Diseases. Antioxidants (Basel) 2021; 10:antiox10060906. [PMID: 34205085 PMCID: PMC8229886 DOI: 10.3390/antiox10060906] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/13/2021] [Accepted: 06/01/2021] [Indexed: 01/09/2023] Open
Abstract
Coenzyme Q10 (CoQ10), which plays a key role in the electron transport chain by providing an adequate, efficient supply of energy, has another relevant function as an antioxidant, acting in mitochondria, other cell compartments, and plasma lipoproteins. CoQ10 deficiency is present in chronic and age-related diseases. In particular, in cardiovascular diseases (CVDs), there is a reduced bioavailability of CoQ10 since statins, one of the most common lipid-lowering drugs, inhibit the common pathway shared by CoQ10 endogenous biosynthesis and cholesterol biosynthesis. Different clinical trials have analyzed the effect of CoQ10 supplementation as a treatment to ameliorate these deficiencies in the context of CVDs. In this review, we focus on recent advances in CoQ10 supplementation and the clinical implications in the reduction of cardiovascular risk factors (such as lipid and lipoprotein levels, blood pressure, or endothelial function) as well as in a therapeutic approach for the reduction of the clinical complications of CVD.
Collapse
Affiliation(s)
- Francisco M. Gutierrez-Mariscal
- Lipids and Atherosclerosis Unit, Unidad de Gestión Clínica de Medicina Interna, Maimonides Institute for Biomedical Research in Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (S.d.l.C.-A.); (J.D.T.-P.); (J.F.A.-D.); (E.M.Y.-S.)
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Silvia de la Cruz-Ares
- Lipids and Atherosclerosis Unit, Unidad de Gestión Clínica de Medicina Interna, Maimonides Institute for Biomedical Research in Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (S.d.l.C.-A.); (J.D.T.-P.); (J.F.A.-D.); (E.M.Y.-S.)
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Jose D. Torres-Peña
- Lipids and Atherosclerosis Unit, Unidad de Gestión Clínica de Medicina Interna, Maimonides Institute for Biomedical Research in Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (S.d.l.C.-A.); (J.D.T.-P.); (J.F.A.-D.); (E.M.Y.-S.)
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Juan F. Alcalá-Diaz
- Lipids and Atherosclerosis Unit, Unidad de Gestión Clínica de Medicina Interna, Maimonides Institute for Biomedical Research in Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (S.d.l.C.-A.); (J.D.T.-P.); (J.F.A.-D.); (E.M.Y.-S.)
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Elena M. Yubero-Serrano
- Lipids and Atherosclerosis Unit, Unidad de Gestión Clínica de Medicina Interna, Maimonides Institute for Biomedical Research in Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (S.d.l.C.-A.); (J.D.T.-P.); (J.F.A.-D.); (E.M.Y.-S.)
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - José López-Miranda
- Lipids and Atherosclerosis Unit, Unidad de Gestión Clínica de Medicina Interna, Maimonides Institute for Biomedical Research in Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (S.d.l.C.-A.); (J.D.T.-P.); (J.F.A.-D.); (E.M.Y.-S.)
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
- Correspondence: ; Tel.: +34-957-012-830
| |
Collapse
|
19
|
Turton N, Bowers N, Khajeh S, Hargreaves IP, Heaton RA. Coenzyme Q10 and the exclusive club of diseases that show a limited response to treatment. Expert Opin Orphan Drugs 2021. [DOI: 10.1080/21678707.2021.1932459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Nadia Turton
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool UK
| | - Nathan Bowers
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool UK
| | - Sam Khajeh
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool UK
| | - Iain P Hargreaves
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool UK
| | - Robert A Heaton
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool UK
| |
Collapse
|
20
|
Rabanal-Ruiz Y, Llanos-González E, Alcain FJ. The Use of Coenzyme Q10 in Cardiovascular Diseases. Antioxidants (Basel) 2021; 10:antiox10050755. [PMID: 34068578 PMCID: PMC8151454 DOI: 10.3390/antiox10050755] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023] Open
Abstract
CoQ10 is an endogenous antioxidant produced in all cells that plays an essential role in energy metabolism and antioxidant protection. CoQ10 distribution is not uniform among different organs, and the highest concentration is observed in the heart, though its levels decrease with age. Advanced age is the major risk factor for cardiovascular disease and endothelial dysfunction triggered by oxidative stress that impairs mitochondrial bioenergetic and reduces NO bioavailability, thus affecting vasodilatation. The rationale of the use of CoQ10 in cardiovascular diseases is that the loss of contractile function due to an energy depletion status in the mitochondria and reduced levels of NO for vasodilatation has been associated with low endogenous CoQ10 levels. Clinical evidence shows that CoQ10 supplementation for prolonged periods is safe, well-tolerated and significantly increases the concentration of CoQ10 in plasma up to 3–5 µg/mL. CoQ10 supplementation reduces oxidative stress and mortality from cardiovascular causes and improves clinical outcome in patients undergoing coronary artery bypass graft surgery, prevents the accumulation of oxLDL in arteries, decreases vascular stiffness and hypertension, improves endothelial dysfunction by reducing the source of ROS in the vascular system and increases the NO levels for vasodilation.
Collapse
Affiliation(s)
- Yoana Rabanal-Ruiz
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071 Ciudad Real, Spain; (Y.R.-R.); (E.L.-G.)
- Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research CRIB, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Emilio Llanos-González
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071 Ciudad Real, Spain; (Y.R.-R.); (E.L.-G.)
- Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research CRIB, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Francisco Javier Alcain
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071 Ciudad Real, Spain; (Y.R.-R.); (E.L.-G.)
- Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research CRIB, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
- Correspondence:
| |
Collapse
|
21
|
Yuan S, Schmidt HM, Wood KC, Straub AC. CoenzymeQ in cellular redox regulation and clinical heart failure. Free Radic Biol Med 2021; 167:321-334. [PMID: 33753238 DOI: 10.1016/j.freeradbiomed.2021.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/22/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
Coenzyme Q (CoQ) is ubiquitously embedded in lipid bilayers of various cellular organelles. As a redox cycler, CoQ shuttles electrons between mitochondrial complexes and extramitochondrial reductases and oxidases. In this way, CoQ is crucial for maintaining the mitochondrial function, ATP synthesis, and redox homeostasis. Cardiomyocytes have a high metabolic rate and rely heavily on mitochondria to provide energy. CoQ levels, in both plasma and the heart, correlate with heart failure in patients, indicating that CoQ is critical for cardiac function. Moreover, CoQ supplementation in clinics showed promising results for treating heart failure. This review provides a comprehensive view of CoQ metabolism and its interaction with redox enzymes and reactive species. We summarize the clinical trials and applications of CoQ in heart failure and discuss the caveats and future directions to improve CoQ therapeutics.
Collapse
Affiliation(s)
- Shuai Yuan
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Heidi M Schmidt
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katherine C Wood
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
22
|
López-Pedrera C, Villalba JM, Patiño-Trives AM, Luque-Tévar M, Barbarroja N, Aguirre MÁ, Escudero-Contreras A, Pérez-Sánchez C. Therapeutic Potential and Immunomodulatory Role of Coenzyme Q 10 and Its Analogues in Systemic Autoimmune Diseases. Antioxidants (Basel) 2021; 10:antiox10040600. [PMID: 33924642 PMCID: PMC8069673 DOI: 10.3390/antiox10040600] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/14/2022] Open
Abstract
Coenzyme Q10 (CoQ10) is a mitochondrial electron carrier and a powerful lipophilic antioxidant located in membranes and plasma lipoproteins. CoQ10 is endogenously synthesized and obtained from the diet, which has raised interest in its therapeutic potential against pathologies related to mitochondrial dysfunction and enhanced oxidative stress. Novel formulations of solubilized CoQ10 and the stabilization of reduced CoQ10 (ubiquinol) have improved its bioavailability and efficacy. Synthetic analogues with increased solubility, such as idebenone, or accumulated selectively in mitochondria, such as MitoQ, have also demonstrated promising properties. CoQ10 has shown beneficial effects in autoimmune diseases. Leukocytes from antiphospholipid syndrome (APS) patients exhibit an oxidative perturbation closely related to the prothrombotic status. In vivo ubiquinol supplementation in APS modulated the overexpression of inflammatory and thrombotic risk-markers. Mitochondrial abnormalities also contribute to immune dysregulation and organ damage in systemic lupus erythematosus (SLE). Idebenone and MitoQ improved clinical and immunological features of lupus-like disease in mice. Clinical trials and experimental models have further demonstrated a therapeutic role for CoQ10 in Rheumatoid Arthritis, multiple sclerosis and type 1 diabetes. This review summarizes the effects of CoQ10 and its analogs in modulating processes involved in autoimmune disorders, highlighting the potential of these therapeutic approaches for patients with immune-mediated diseases.
Collapse
Affiliation(s)
- Chary López-Pedrera
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
- Correspondence: ; Tel.: +34-957-213795
| | - José Manuel Villalba
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, 14014 Córdoba, Spain; (J.M.V.); (C.P.-S.)
| | - Alejandra Mª Patiño-Trives
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Maria Luque-Tévar
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Nuria Barbarroja
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Mª Ángeles Aguirre
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Alejandro Escudero-Contreras
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Carlos Pérez-Sánchez
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, 14014 Córdoba, Spain; (J.M.V.); (C.P.-S.)
| |
Collapse
|
23
|
The stabilization and antioxidant performances of coenzyme Q10-loaded niosomes coated by PEG and chitosan. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2020.115194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
24
|
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: 2.5] [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
|
25
|
Pastor-Maldonado CJ, Suárez-Rivero JM, Povea-Cabello S, Álvarez-Córdoba M, Villalón-García I, Munuera-Cabeza M, Suárez-Carrillo A, Talaverón-Rey M, Sánchez-Alcázar JA. Coenzyme Q 10: Novel Formulations and Medical Trends. Int J Mol Sci 2020; 21:E8432. [PMID: 33182646 PMCID: PMC7697799 DOI: 10.3390/ijms21228432] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 10/31/2020] [Accepted: 11/07/2020] [Indexed: 12/11/2022] Open
Abstract
The aim of this review is to shed light over the most recent advances in Coenzyme Q10 (CoQ10) applications as well as to provide detailed information about the functions of this versatile molecule, which have proven to be of great interest in the medical field. Traditionally, CoQ10 clinical use was based on its antioxidant properties; however, a wide range of highly interesting alternative functions have recently been discovered. In this line, CoQ10 has shown pain-alleviating properties in fibromyalgia patients, a membrane-stabilizing function, immune system enhancing ability, or a fundamental role for insulin sensitivity, apart from potentially beneficial properties for familial hypercholesterolemia patients. In brief, it shows a remarkable amount of functions in addition to those yet to be discovered. Despite its multiple therapeutic applications, CoQ10 is not commonly prescribed as a drug because of its low oral bioavailability, which compromises its efficacy. Hence, several formulations have been developed to face such inconvenience. These were initially designed as lipid nanoparticles for CoQ10 encapsulation and distribution through biological membranes and eventually evolved towards chemical modifications of the molecule to decrease its hydrophobicity. Some of the most promising formulations will also be discussed in this review.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - José A. Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III. Universidad Pablo de Olavide, 41013 Sevilla, Spain; (C.J.P.-M.); (J.M.S.-R.); (S.P.-C.); (M.Á.-C.); (I.V.-G.); (M.M.-C.); (A.S.-C.); (M.T.-R.)
| |
Collapse
|
26
|
Gutierrez-Mariscal FM, Arenas-de Larriva AP, Limia-Perez L, Romero-Cabrera JL, Yubero-Serrano EM, López-Miranda J. Coenzyme Q 10 Supplementation for the Reduction of Oxidative Stress: Clinical Implications in the Treatment of Chronic Diseases. Int J Mol Sci 2020; 21:ijms21217870. [PMID: 33114148 PMCID: PMC7660335 DOI: 10.3390/ijms21217870] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/15/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Apart from its main function in the mitochondria as a key element in electron transport, Coenzyme Q10 (CoQ10) has been described as having multiple functions, such as oxidant action in the generation of signals and the control of membrane structure and phospholipid and cellular redox status. Among these, the most relevant and most frequently studied function is the potent antioxidant capability of its coexistent redox forms. Different clinical trials have investigated the effect of CoQ10 supplementation and its ability to reduce oxidative stress. In this review, we focused on recent advances in CoQ10 supplementation, its role as an antioxidant, and the clinical implications that this entails in the treatment of chronic diseases, in particular cardiovascular diseases, kidney disease, chronic obstructive pulmonary disease, non-alcoholic fatty liver disease, and neurodegenerative diseases. As an antioxidant, CoQ10 has proved to be of potential use as a treatment in diseases in which oxidative stress is a hallmark, and beneficial effects of CoQ10 have been reported in the treatment of chronic diseases. However, it is crucial to reach a consensus on the optimal dose and the use of different formulations, which vary from ubiquinol or ubiquinone Ubisol-Q10 or Qter®, to new analogues such as MitoQ, before we can draw a clear conclusion about its clinical use. In addition, a major effort must be made to demonstrate its beneficial effects in clinical trials, with a view to making the implementation of CoQ10 possible in clinical practice.
Collapse
Affiliation(s)
- Francisco Miguel Gutierrez-Mariscal
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Córdoba, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (A.P.A.-d.L.); (L.L.-P.); (J.L.R.-C.)
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Antonio Pablo Arenas-de Larriva
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Córdoba, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (A.P.A.-d.L.); (L.L.-P.); (J.L.R.-C.)
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Laura Limia-Perez
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Córdoba, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (A.P.A.-d.L.); (L.L.-P.); (J.L.R.-C.)
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Juan Luis Romero-Cabrera
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Córdoba, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (A.P.A.-d.L.); (L.L.-P.); (J.L.R.-C.)
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Elena Maria Yubero-Serrano
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Córdoba, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (A.P.A.-d.L.); (L.L.-P.); (J.L.R.-C.)
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Correspondence: (E.M.Y.-S.); (J.L.-M.); Tel.: +34-957213733 (E.M.Y.-S.); +34-957010947 (J.L.-M.); Fax: +34-957218250 (J.L.-M.)
| | - Jose López-Miranda
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Córdoba, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; (F.M.G.-M.); (A.P.A.-d.L.); (L.L.-P.); (J.L.R.-C.)
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Correspondence: (E.M.Y.-S.); (J.L.-M.); Tel.: +34-957213733 (E.M.Y.-S.); +34-957010947 (J.L.-M.); Fax: +34-957218250 (J.L.-M.)
| |
Collapse
|
27
|
Pshenichnyuk SA, Modelli A, Asfandiarov NL, Komolov AS. Ionizing radiation and natural constituents of living cells: Low-energy electron interaction with coenzyme Q analogs. J Chem Phys 2020; 153:111103. [PMID: 32962391 DOI: 10.1063/5.0022188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Resonance electron attachment to short-tail analogs of coenzyme Q10 is investigated in the electron energy range 0 eV-14 eV under gas-phase conditions by means of dissociative electron attachment spectroscopy. Formation of long-lived (milliseconds) molecular negative ions is detected at 1.2 eV, but not at thermal energy. A huge increase in the electron detachment time as compared with the reference para-benzoquinone (40 µs) is ascribed to the presence of the isoprene side chains. Elimination of a neutral CH3 radical is found to be the most intense decay detected on the microsecond time scale. The results give some insight into the timescale of electron-driven processes stimulated in living tissues by high-energy radiation and are of importance in prospective fields of radiobiology and medicine.
Collapse
Affiliation(s)
- Stanislav A Pshenichnyuk
- Institute of Molecule and Crystal Physics, Ufa Federal Research Centre, Russian Academy of Sciences, Prospekt Oktyabrya 151, 450075 Ufa, Russia
| | - Alberto Modelli
- Università di Bologna, Dipartimento di Chimica "G. Ciamician", Via Selmi 2, 40126 Bologna, Italy
| | - Nail L Asfandiarov
- Institute of Molecule and Crystal Physics, Ufa Federal Research Centre, Russian Academy of Sciences, Prospekt Oktyabrya 151, 450075 Ufa, Russia
| | - Alexey S Komolov
- St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia
| |
Collapse
|
28
|
Rajdev K, Mehan S. Neuroprotective Methodologies of Co-Enzyme Q10 Mediated Brain Hemorrhagic Treatment: Clinical and Pre-Clinical Findings. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 18:446-465. [PMID: 31187715 DOI: 10.2174/1871527318666190610101144] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 04/22/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022]
Abstract
Cerebral brain hemorrhage is associated with the highest mortality and morbidity despite only constituting approximately 10-15% of all strokes classified into intracerebral and intraventricular hemorrhage where most of the patients suffer from impairment in memory, weakness or paralysis in arms or legs, headache, fatigue, gait abnormality and cognitive dysfunctions. Understanding molecular pathology and finding the worsening cause of hemorrhage will lead to explore the therapeutic interventions that could prevent and cure the disease. Mitochondrial ETC-complexes dysfunction has been found to increase neuroinflammatory cytokines, oxidative free radicals, excitotoxicity, neurotransmitter and energy imbalance that are the key neuropathological hallmarks of cerebral hemorrhage. Coenzyme Q10 (CoQ10), as a part of the mitochondrial respiratory chain can effectively restore these neuronal dysfunctions by preventing the opening of mitochondrial membrane transition pore, thereby counteracting cell death events as well as exerts an anti-inflammatory effect by influencing the expression of NF-kB1 dependent genes thus preventing the neuroinflammation and energy restoration. Due to behavior and biochemical heterogeneity in post cerebral brain hemorrhagic pattern different preclinical autologous blood injection models are required to precisely investigate the forthcoming therapeutic strategies. Despite emerging pre-clinical research and resultant large clinical trials for promising symptomatic treatments, there are very less pharmacological interventions demonstrated to improve post operative condition of patients where intensive care is required. Therefore, in current review, we explore the disease pattern, clinical and pre-clinical interventions under investigation and neuroprotective methodologies of CoQ10 precursors to ameliorate post brain hemorrhagic conditions.
Collapse
Affiliation(s)
- Kajal Rajdev
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Sidharth Mehan
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| |
Collapse
|
29
|
Manolis AS, Manolis AA, Manolis TA, Apostolaki NE, Apostolopoulos EJ, Melita H, Katsiki N. Mitochondrial dysfunction in cardiovascular disease: Current status of translational research/clinical and therapeutic implications. Med Res Rev 2020; 41:275-313. [PMID: 32959403 DOI: 10.1002/med.21732] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022]
Abstract
Mitochondria provide energy to the cell during aerobic respiration by supplying ~95% of the adenosine triphosphate (ATP) molecules via oxidative phosphorylation. These organelles have various other functions, all carried out by numerous proteins, with the majority of them being encoded by nuclear DNA (nDNA). Mitochondria occupy ~1/3 of the volume of myocardial cells in adults, and function at levels of high-efficiency to promptly meet the energy requirements of the myocardial contractile units. Mitochondria have their own DNA (mtDNA), which contains 37 genes and is maternally inherited. Over the last several years, a variety of functions of these organelles have been discovered and this has led to a growing interest in their involvement in various diseases, including cardiovascular (CV) diseases. Mitochondrial dysfunction relates to the status where mitochondria cannot meet the demands of a cell for ATP and there is an enhanced formation of reactive-oxygen species. This dysfunction may occur as a result of mtDNA and/or nDNA mutations, but also as a response to aging and various disease and environmental stresses, leading to the development of cardiomyopathies and other CV diseases. Designing mitochondria-targeted therapeutic strategies aiming to maintain or restore mitochondrial function has been a great challenge as a result of variable responses according to the etiology of the disorder. There have been several preclinical data on such therapies, but clinical studies are scarce. A major challenge relates to the techniques needed to eclectically deliver the therapeutic agents to cardiac tissues and to damaged mitochondria for successful clinical outcomes. All these issues and progress made over the last several years are herein reviewed.
Collapse
Affiliation(s)
- Antonis S Manolis
- First Department of Cardiology, Athens University School of Medicine, Athens, Greece
| | | | | | | | | | | | - Niki Katsiki
- First Department of Internal Medicine, Division of Endocrinology and Metabolism, Diabetes Center, Medical School, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
30
|
Park HW, Park CG, Park M, Lee SH, Park HR, Lim J, Paek SH, Choy YB. Intrastriatal administration of coenzyme Q10 enhances neuroprotection in a Parkinson's disease rat model. Sci Rep 2020; 10:9572. [PMID: 32533070 PMCID: PMC7293316 DOI: 10.1038/s41598-020-66493-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease is a neurodegenerative disorder, and no treatment has been yet established to prevent disease progression. Coenzyme Q10, an antioxidant, has been considered a promising neuroprotective agent; however, conventional oral administration provides limited efficacy due to its very low bioavailability. In this study, we hypothesised that continuous, intrastriatal administration of a low dose of Coenzyme Q10 could effectively prevent dopaminergic neuron degeneration. To this end, a Parkinson's disease rat model induced by 6-hydroxydopamine was established, and the treatment was applied a week before the full establishment of this disease model. Behavioural tests showed a dramatically decreased number of asymmetric rotations in the intrastriatal Coenzyme Q10 group compared with the no treatment group. Rats with intrastriatal Coenzyme Q10 exposure also exhibited a larger number of dopaminergic neurons, higher expression of neurogenetic and angiogenetic factors, and less inflammation, and the effects were more prominent than those of orally administered Coenzyme Q10, although the dose of intrastriatal Coenzyme Q10 was 17,000-times lower than that of orally-administered Coenzyme Q10. Therefore, continuous, intrastriatal delivery of Coenzyme Q10, especially when combined with implantable devices for convection-enhanced delivery or deep brain stimulation, can be an effective strategy to prevent neurodegeneration in Parkinson's disease.
Collapse
Affiliation(s)
- Hyung Woo Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Republic of Korea
| | - Min Park
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Ho Lee
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hye Ran Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jaesung Lim
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Young Bin Choy
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul, 03080, Republic of Korea.
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
31
|
Ryu KA, Park PJ, Kim SB, Bin BH, Jang DJ, Kim ST. Topical Delivery of Coenzyme Q10-Loaded Microemulsion for Skin Regeneration. Pharmaceutics 2020; 12:pharmaceutics12040332. [PMID: 32272811 PMCID: PMC7238272 DOI: 10.3390/pharmaceutics12040332] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/01/2020] [Accepted: 04/04/2020] [Indexed: 02/07/2023] Open
Abstract
The aim of this study was to develop a coenzyme Q10 (CoQ10) microemulsion system with improved solubility, penetration, and wound healing efficacy. Based on the pseudo-ternary diagram, microemulsions containing isopropyl myristate (IPM), Cremophor EL®, and Transcutol® HP were selected and confirmed to be nanosized (<20 nm) and thermodynamically stable based on the dilution and thermodynamic stability tests. The CoQ10-loaded microemulsion with a surfactant/co-surfactant (S/CoS) ratio of 2:1 (w/w %) demonstrated a higher permeation efficacy compared to microemulsions with S/CoS ratio of 3:1 or 4:1 (w/w %). Additionally, the CoQ10-loaded microemulsion with an S/CoS ratio of 2:1 demonstrated a relatively rapid wound healing effect in keratinocytes and fibroblasts. Overall, these data suggest that a microemulsion based on IPM, Cremophor EL®, and Transcutol® HP could be an effective vehicle for the topical administration of CoQ10 and could be utilized for the application of other therapeutic agents that have difficulty in penetrating the skin.
Collapse
Affiliation(s)
- Kyeong-A Ryu
- Institute of Digital Anti-Aging Healthcare, Inje University, Gimhae 50834, Korea;
| | - Phil June Park
- Department of Genetic Engineering, Sungkyunkwan University, Suwon-si, Gyeonggi-do 16499, Korea;
| | - Seong-Bo Kim
- Bio-Living Engineering Major, Global Leaders College, Yonsei University, 50 yonsei-ro, Shinchon-dong, Seodaemun-gu, Seoul 03722, Korea;
| | - Bum-Ho Bin
- Department of Biological Sciences, Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Korea;
| | - Dong-Jin Jang
- Institute of Digital Anti-Aging Healthcare, Inje University, Gimhae 50834, Korea;
- Department of Pharmaceutical Engineering, Inje University, Gimhae 50834, Korea
- Correspondence: (D.-J.J.); (S.T.K.); Tel.: +82-55-320-3393 (D.-J.J.); +82-55-320-4038 (S.T.K.)
| | - Sung Tae Kim
- Institute of Digital Anti-Aging Healthcare, Inje University, Gimhae 50834, Korea;
- Department of Pharmaceutical Engineering, Inje University, Gimhae 50834, Korea
- Correspondence: (D.-J.J.); (S.T.K.); Tel.: +82-55-320-3393 (D.-J.J.); +82-55-320-4038 (S.T.K.)
| |
Collapse
|
32
|
Ibrahim Fouad G. Combination of Omega 3 and Coenzyme Q10 Exerts Neuroprotective Potential Against Hypercholesterolemia-Induced Alzheimer's-Like Disease in Rats. Neurochem Res 2020; 45:1142-1155. [PMID: 32124160 DOI: 10.1007/s11064-020-02996-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/15/2020] [Accepted: 02/20/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia that progressively disrupts neurocognitive function, which has neither cure nor effective treatment. Hypercholesterolemia might be involved in brain alterations that could evolve into AD. The present study aims to evaluate the potential of omega-3, Co-enzyme Q10 (Co-Q10), as well as their combination in ameliorating hypercholesterolemia-initiated AD-like disease. We adapted a hypercholesterolemic (HC) rat model, a model of oxidative stress-mediated neurodegeneration, to study AD-like pathology. Hypercholesterolemia resulted in increased lipid peroxidation coupled with declined nitric oxide production, reduced glutathione levels, and decreased antioxidant activities of glutathione-s-transferase (GST) and glutathione peroxidase (GSH-Px) in the brain. Moreover, hypercholesterolemia resulted in decreased acetylcholine (ACh) levels and increased acetylcholine-esterase (AChE) activity, along with an increment of tumor necrosis factor and amyloid-β 42. Behaviorally, HC-rats demonstrated depressive-like behavior and declined memory. Treatment of HC-rats with omega-3 and Co-Q10 (alone or in combination) alleviated the brain oxidative stress and inflammation, regulated cholinergic functioning, and enhanced the functional outcome. These findings were verified by the histopathological investigation of brain tissues. This neuroprotective potential of omega-3 and Co-Q10 was achieved through anti-oxidative, anti-inflammatory, anti-amyloidogenic, pro-cholinergic, and memory-enhancing activities against HC-induced AD-like disease; suggesting that they may be useful as prophylactic and therapeutic agents against the neurotoxic effects of hypercholesterolemia.
Collapse
Affiliation(s)
- Ghadha Ibrahim Fouad
- Department of Therapeutic Chemistry, National Research Centre, 33 El-Bohouth St., Dokki, Cairo, 12622, Egypt.
| |
Collapse
|
33
|
Liu Z, Liu X, Yang Q, Yu L, Chang Y, Qu M. Neutrophil membrane-enveloped nanoparticles for the amelioration of renal ischemia-reperfusion injury in mice. Acta Biomater 2020; 104:158-166. [PMID: 31954188 DOI: 10.1016/j.actbio.2020.01.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 12/21/2022]
Abstract
Ischemia-reperfusion (I/R) injury initiates and exacerbates a series of oxidative and inflammatory events, and causes high morbidity and mortality. Despite the progress made with recent clinical use of anti-malarial drugs, the response rate of I/R injury treatment remains unsatisfactory. Here, we showed a neutrophil membrane-enveloped Coenzyme Q (N-NPCoQ10) nanoparticle strategy for I/R injury treatment. We validated the physicochemical and biological reproducibility of the nanoparticles and tested the protective effects of N-NPCoQ10 in oxygen-glucose deprivation/reperfusion model and renal I/R injury mouse model. N-NPCoQ10 nanoparticles administration exhibited synergistic protective effect against I/R injury, which significantly reduced oxidative damage in vitro and in vivo, inhibited renal cell apoptosis, attenuated inflammatory response in renal I/R injury model, and finally improved renal function of I/R injury mice. The N-NPCoQ10 nanoparticles administration provides an efficient way to deliver anti-oxidant that suppresses oxidative damages and neutralize proinflammatory cytokines during renal I/R injury, which might be a potential strategy for renal acute kidney injury treatment. STATEMENT OF SIGNIFICANCE: The neutrophil membrane-enveloped Coenzyme Q nanoparticles (N-NPCoQ10) provides an efficient way to protect oxidative, inflammatory, and apoptotic reaction in renal I/R injury, which might be a potential strategy for renal acute kidney injury treatment.
Collapse
|
34
|
Moschetti A, Vine LN, Lethcoe K, Dagda RK, Ellison P, Ryan RO. Assembly and Characterization of Biocompatible Coenzyme Q 10 -Enriched Lipid Nanoparticles. Lipids 2020; 55:141-149. [PMID: 32074388 DOI: 10.1002/lipd.12218] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/18/2019] [Accepted: 01/17/2020] [Indexed: 12/18/2022]
Abstract
Coenzyme Q10 (CoQ10 ) is a strongly hydrophobic lipid that functions in the electron transport chain and as an antioxidant. CoQ10 was conferred with aqueous solubility by incorporation into nanoparticles containing phosphatidylcholine (PtdCho) and apolipoprotein (apo) A-I. These particles, termed CoQ10 nanodisks (ND), contain 1.0 mg CoQ10 /5 mg PtdCho/2 mg apoA-I (97% CoQ10 solubilization efficiency). UV/Vis absorbance spectroscopy of CoQ10 ND revealed a characteristic absorbance peak centered at 275 nm. Incorporation of CoQ10 into ND resulted in quenching of apoA-I tryptophan fluorescence emission. Gel filtration chromatography of CoQ10 ND gave rise to a single major absorbance peak and HPLC of material extracted from this peak confirmed the presence of CoQ10 . Incubation of cultured cells with CoQ10 ND, but not empty ND, resulted in a significant increase in the CoQ10 content of mitochondria as well as enhanced oxidative phosphorylation, as observed by a ~24% increase in maximal oxygen consumption rate. Collectively, a facile method to solubilize significant quantities of CoQ10 in lipid nanoparticles has been developed. The availability of CoQ10 ND provides a novel means to investigate biochemical aspects of CoQ10 uptake by cells and/or administer it to subjects deficient in this key lipid as a result of inborn errors of metabolism, statin therapy, or otherwise.
Collapse
Affiliation(s)
- Anthony Moschetti
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, 89557, USA
| | - Lucas N Vine
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, 89557, USA
| | - Kyle Lethcoe
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, 89557, USA
| | - Ruben K Dagda
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Patricia Ellison
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, 89557, USA
| | - Robert O Ryan
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV, 89557, USA
| |
Collapse
|
35
|
Arenas‐Jal M, Suñé‐Negre JM, García‐Montoya E. Coenzyme Q10 supplementation: Efficacy, safety, and formulation challenges. Compr Rev Food Sci Food Saf 2020; 19:574-594. [DOI: 10.1111/1541-4337.12539] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/26/2019] [Accepted: 01/03/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Marta Arenas‐Jal
- Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy and Food SciencesUniversity of Barcelona Barcelona Spain
| | - J. M. Suñé‐Negre
- Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy and Food SciencesUniversity of Barcelona Barcelona Spain
| | - Encarna García‐Montoya
- Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy and Food SciencesUniversity of Barcelona Barcelona Spain
| |
Collapse
|
36
|
Serag H, El Wakeel L, Adly A. Coenzyme Q10 administration has no effect on sICAM-1 and metabolic parameters of pediatrics with type 1 diabetes mellitus. INT J VITAM NUTR RES 2020; 91:315-324. [PMID: 31942840 DOI: 10.1024/0300-9831/a000636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background: Endothelial dysfunction (ED) plays a key role in the development and progression of microvascular and macrovascular complications in pediatrics with type 1 diabetes mellitus (T1DM). Coenzyme Q10 (CoQ10) is a nutraceutical with a known anti-inflammatory and anti-oxidant activity. This study was conducted to evaluate the potential effect of CoQ10 on ED and various metabolic parameters. Methods: This prospective randomized open-label pilot study was conducted on 49 T1DM pediatric patients. Seven healthy non-diabetic pediatric subjects who didn't receive treatment were included as a control group. Eligible patients were randomly allocated into either group I (n = 25); received 100 mg of CoQ10 in addition to standard treatment or group II (n = 24); received standard treatment only. The levels of; soluble intracellular adhesion molecule-1 (sICAM-1), glycated hemoglobin (HbA1c), fasting blood glucose (FBG), lipid profile, serum creatinine and liver function tests were assessed for both groups at baseline and after 3 months of treatment. Results: At baseline, compared to an age-matched healthy control group sICAM-1 levels were significantly elevated in group II diabetic patients (276.5 (231.6-320.66) vs 221.8 (177.9-267.1 ng/ml), p = 0.042. After 3 months of treatment no significant difference was observed in sICAM-1, HbA1c, FBG, lipid profile, serum creatinine and liver function tests between the two study groups. A positive correlation was found between sICAM-1 and HbA1c throughout the study (r = 0.308, p = 0.0054). Conclusion: Administration of CoQ10 for 3 months in T1DM pediatric patients was well tolerated but had no favorable effect on ED or metabolic parameters.
Collapse
Affiliation(s)
- Heba Serag
- Department of Clinical Pharmacy, Faculty of Pharmacy Ain Shams University, Cairo, Egypt
| | - Lamia El Wakeel
- Department of Clinical Pharmacy, Faculty of Pharmacy Ain Shams University, Cairo, Egypt
| | - Amira Adly
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
37
|
Sarafian TA, Yacoub A, Kunz A, Aranki B, Serobyan G, Cohn W, Whitelegge JP, Watson JB. Enhanced mitochondrial inhibition by 3,4-dihydroxyphenyl-acetaldehyde (DOPAL)-oligomerized α-synuclein. J Neurosci Res 2019; 97:1689-1705. [PMID: 31420910 DOI: 10.1002/jnr.24513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/24/2019] [Accepted: 07/23/2019] [Indexed: 12/29/2022]
Abstract
Oligomeric forms of α-synuclein are believed to cause mitochondrial injury, which may contribute to neurotoxicity in Parkinson's disease (PD). Here oligomers of α-synuclein were prepared using the dopamine metabolite, DOPAL (3,4-dihydroxyphenyl-acetaldehyde), in the presence of guanidinium hydrochloride. Electron microscopy, mass spectrometry, and Western blotting studies revealed enhanced and stable oligomerization with DOPAL compared with dopamine or CuCl2 /H2 O2 . Using isolated mouse brain mitochondria, DOPAL-oligomerized α-synuclein (DOS) significantly inhibited oxygen consumption rates compared with untreated, control-fibrillated, and dopamine-fibrillated synuclein, or with monomeric α-synuclein. Inhibition was greater in the presence of malate plus pyruvate than with succinate, suggesting the involvement of mitochondrial complex I. Mitochondrial membrane potential studies using fluorescent probes, JC-1, and Safranin O also detected enhanced inhibition by DOS compared with the other aggregated forms of α-synuclein. Testing a small customized chemical library, four compounds were identified that rescued membrane potential from DOS injury. While diverse in chemical structure and mechanism, each compound has been reported to interact with mitochondrial complex I. Western blotting studies revealed that none of the four compounds disrupted the oligomeric banding pattern of DOS, suggesting their protection involved direct mitochondrial interaction. The remaining set of chemicals also did not disrupt oligomeric banding, attesting to the high structural stability of this α-synuclein proteoform. DOPAL and α-synuclein are both found in dopaminergic neurons, where their levels are elevated in PD and in animal models exposed to chemical toxicants, including agricultural pesticides. The current study provides further evidence of α-synuclein-induced mitochondrial injury and a likely role in PD neuropathology.
Collapse
Affiliation(s)
- Theodore A Sarafian
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Amneh Yacoub
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Anastasia Kunz
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Burkan Aranki
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Grigor Serobyan
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Whitaker Cohn
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Julian P Whitelegge
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| | - Joseph B Watson
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California
| |
Collapse
|
38
|
Cerqua C, Casarin A, Pierrel F, Vazquez Fonseca L, Viola G, Salviati L, Trevisson E. Vitamin K2 cannot substitute Coenzyme Q 10 as electron carrier in the mitochondrial respiratory chain of mammalian cells. Sci Rep 2019; 9:6553. [PMID: 31024065 PMCID: PMC6484000 DOI: 10.1038/s41598-019-43014-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/11/2019] [Indexed: 12/16/2022] Open
Abstract
Coenzyme Q10 (CoQ10) deficiencies are a group of heterogeneous conditions that respond to ubiquinone administration if treated soon after the onset of symptoms. However, this treatment is only partially effective due to its poor bioavailability. We tested whether vitamin K2, which was reported to act as a mitochondrial electron carrier in D. melanogaster, could mimic ubiquinone function in human CoQ10 deficient cell lines, and in yeast carrying mutations in genes required for coenzyme Q6 (CoQ6) biosynthesis. We found that vitamin K2, despite entering into mitochondria, restored neither electron flow in the respiratory chain, nor ATP synthesis. Conversely, coenzyme Q4 (CoQ4), an analog of CoQ10 with a shorter isoprenoid side chain, could efficiently substitute its function. Given its better solubility, CoQ4 could represent an alternative to CoQ10 in patients with both primary and secondary CoQ10 deficiencies.
Collapse
Affiliation(s)
- Cristina Cerqua
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy.,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy
| | - Alberto Casarin
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy.,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy
| | - Fabien Pierrel
- Univ. Grenoble Alpes, CNRS, CHU Grenoble Alpes, Grenoble INP, TIMC-IMAG, 38000, Grenoble, France
| | - Luis Vazquez Fonseca
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy.,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy
| | - Giampiero Viola
- Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.,Pediatric Hematooncology Laboratory, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy
| | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy. .,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.
| | - Eva Trevisson
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy. .,Istituto di Ricerca Pediatrica IRP Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.
| |
Collapse
|
39
|
Mahfoz AM. Renal Protective Effects of Coenzyme Q10 Against Chromate Induced Nephrotoxicity in Rats. ACTA ACUST UNITED AC 2019. [DOI: 10.3923/jas.2019.453.458] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
40
|
Kunzelmann K, Ousingsawat J, Cabrita I, Doušová T, Bähr A, Janda M, Schreiber R, Benedetto R. TMEM16A in Cystic Fibrosis: Activating or Inhibiting? Front Pharmacol 2019; 10:3. [PMID: 30761000 PMCID: PMC6362895 DOI: 10.3389/fphar.2019.00003] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/04/2019] [Indexed: 12/26/2022] Open
Abstract
The inflammatory airway disease cystic fibrosis (CF) is characterized by airway obstruction due to mucus hypersecretion, airway plugging, and bronchoconstriction. The cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel is dysfunctional in CF, leading to defects in epithelial transport. Although CF pathogenesis is still disputed, activation of alternative Cl- channels is assumed to improve lung function in CF. Two suitable non-CFTR Cl- channels are present in the airway epithelium, the Ca2+ activated channel TMEM16A and SLC26A9. Activation of these channels is thought to be feasible to improve hydration of the airway mucus and to increase mucociliary clearance. Interestingly, both channels are upregulated during inflammatory lung disease. They are assumed to support fluid secretion, necessary to hydrate excess mucus and to maintain mucus clearance. During inflammation, however, TMEM16A is upregulated particularly in mucus producing cells, with only little expression in ciliated cells. Recently it was shown that knockout of TMEM16A in ciliated cells strongly compromises Cl- conductance and attenuated mucus secretion, but does not lead to a CF-like lung disease and airway plugging. Along this line, activation of TMEM16A by denufosol, a stable purinergic ligand, failed to demonstrate any benefit to CF patients in earlier studies. It rather induced adverse effects such as cough. A number of studies suggest that TMEM16A is essential for mucus secretion and possibly also for mucus production. Evidence is now provided for a crucial role of TMEM16A in fusion of mucus-filled granules with the apical plasma membrane and cellular exocytosis. This is probably due to local Ca2+ signals facilitated by TMEM16A. Taken together, TMEM16A supports fluid secretion by ciliated airway epithelial cells, but also maintains excessive mucus secretion during inflammatory airway disease. Because TMEM16A also supports airway smooth muscle contraction, inhibition rather than activation of TMEM16A might be the appropriate treatment for CF lung disease, asthma and COPD. As a number of FDA-approved and well-tolerated drugs have been shown to inhibit TMEM16A, evaluation in clinical trials appears timely.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | | | - Inês Cabrita
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Tereza Doušová
- Department of Pediatrics, Second Faculty of Medicine, University Hospital Motol, Charles University in Prague, Prague, Czechia
| | - Andrea Bähr
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, Munich, Germany
- Innere Medizin I, Klinikum Rechts der Isar der TU München, München, Germany
| | - Melanie Janda
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Roberta Benedetto
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| |
Collapse
|
41
|
Frontiñán-Rubio J, Sancho-Bielsa FJ, Peinado JR, LaFerla FM, Giménez-Llort L, Durán-Prado M, Alcain FJ. Sex-dependent co-occurrence of hypoxia and β-amyloid plaques in hippocampus and entorhinal cortex is reversed by long-term treatment with ubiquinol and ascorbic acid in the 3 × Tg-AD mouse model of Alzheimer's disease. Mol Cell Neurosci 2018; 92:67-81. [PMID: 29953929 DOI: 10.1016/j.mcn.2018.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 12/15/2022] Open
Abstract
Structural and functional abnormalities in the cerebral microvasculature have been observed in Alzheimer's disease (AD) patients and animal models. One cause of hypoperfusion is the thickening of the cerebrovascular basement membrane (CVBM) due to increased collagen-IV deposition around capillaries. This study investigated whether these and other alterations in the cerebrovascular system associated with AD can be prevented by long-term dietary supplementation with the antioxidant ubiquinol (Ub) stabilized with Kaneka QH P30 powder containing ascorbic acid (ASC) in a mouse model of advanced AD (3 × Tg-AD mice, 12 months old). Animals were treated from prodromal stages of disease (3 months of age) with standard chow without or with Ub + ASC or ASC-containing vehicle and compared to wild-type (WT) mice. The number of β-amyloid (Aβ) plaques in the hippocampus and entorhinal cortex was higher in female than in male 3 × Tg-AD mice. Extensive regions of hypoxia were characterized by a higher plaque burden in females only. This was abolished by Ub + ASC and, to a lesser extent, by ASC treatment. Irrespective of Aβ burden, increased collagen-IV deposition in the CVBM was observed in both male and female 3 × Tg-AD mice relative to WT animals; this was also abrogated in Ub + ASC- and ASC-treated mice. The chronic inflammation in the hippocampus and oxidative stress in peripheral leukocytes of 3 × Tg-AD mice were likewise reversed by antioxidant treatment. These results provide strong evidence that long-term antioxidant treatment can mitigate plasma oxidative stress, amyloid burden, and hypoxia in the AD brain parenchyma.
Collapse
Affiliation(s)
- Javier Frontiñán-Rubio
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Spain
| | - Francisco J Sancho-Bielsa
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Spain
| | - Juan R Peinado
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Spain
| | - Frank M LaFerla
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Lydia Giménez-Llort
- Department of Psychiatry and Forensic Medicine, Faculty of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain; Institut of Neuroscience, Faculty of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Mario Durán-Prado
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Spain.
| | - Francisco J Alcain
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Spain.
| |
Collapse
|
42
|
Giorgi C, Marchi S, Simoes IC, Ren Z, Morciano G, Perrone M, Patalas-Krawczyk P, Borchard S, Jȩdrak P, Pierzynowska K, Szymański J, Wang DQ, Portincasa P, Wȩgrzyn G, Zischka H, Dobrzyn P, Bonora M, Duszynski J, Rimessi A, Karkucinska-Wieckowska A, Dobrzyn A, Szabadkai G, Zavan B, Oliveira PJ, Sardao VA, Pinton P, Wieckowski MR. Mitochondria and Reactive Oxygen Species in Aging and Age-Related Diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 340:209-344. [PMID: 30072092 PMCID: PMC8127332 DOI: 10.1016/bs.ircmb.2018.05.006] [Citation(s) in RCA: 248] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aging has been linked to several degenerative processes that, through the accumulation of molecular and cellular damage, can progressively lead to cell dysfunction and organ failure. Human aging is linked with a higher risk for individuals to develop cancer, neurodegenerative, cardiovascular, and metabolic disorders. The understanding of the molecular basis of aging and associated diseases has been one major challenge of scientific research over the last decades. Mitochondria, the center of oxidative metabolism and principal site of reactive oxygen species (ROS) production, are crucial both in health and in pathogenesis of many diseases. Redox signaling is important for the modulation of cell functions and several studies indicate a dual role for ROS in cell physiology. In fact, high concentrations of ROS are pathogenic and can cause severe damage to cell and organelle membranes, DNA, and proteins. On the other hand, moderate amounts of ROS are essential for the maintenance of several biological processes, including gene expression. In this review, we provide an update regarding the key roles of ROS-mitochondria cross talk in different fundamental physiological or pathological situations accompanying aging and highlighting that mitochondrial ROS may be a decisive target in clinical practice.
Collapse
Affiliation(s)
- Carlotta Giorgi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Saverio Marchi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Ines C.M. Simoes
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ziyu Ren
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Giampaolo Morciano
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
- Maria Pia Hospital, GVM Care & Research, Torino, Italy
| | - Mariasole Perrone
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paulina Patalas-Krawczyk
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Sabine Borchard
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Paulina Jȩdrak
- Department of Molecular Biology, University of Gdańsk, Gdańsk, Poland
| | | | - Jȩdrzej Szymański
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - David Q. Wang
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Dept. of Biomedical Sciences & Human Oncology, University of Bari "Aldo Moro" Medical School, Bari, Italy
| | - Grzegorz Wȩgrzyn
- Department of Molecular Biology, University of Gdańsk, Gdańsk, Poland
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, Munich, Germany
| | - Pawel Dobrzyn
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Massimo Bonora
- Departments of Cell Biology and Gottesman Institute for Stem Cell & Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jerzy Duszynski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Alessandro Rimessi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | | | | | - Gyorgy Szabadkai
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Barbara Zavan
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Paulo J. Oliveira
- CNC - Center for Neuroscience and Cell Biology, UC-Biotech, Biocant Park, University of Coimbra, Cantanhede, Portugal
| | - Vilma A. Sardao
- CNC - Center for Neuroscience and Cell Biology, UC-Biotech, Biocant Park, University of Coimbra, Cantanhede, Portugal
| | - Paolo Pinton
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
| | - Mariusz R. Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
43
|
Gutierrez-Mariscal FM, Yubero-Serrano EM, Villalba JM, Lopez-Miranda J. Coenzyme Q10: From bench to clinic in aging diseases, a translational review. Crit Rev Food Sci Nutr 2018; 59:2240-2257. [DOI: 10.1080/10408398.2018.1442316] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Francisco M. Gutierrez-Mariscal
- Lipid and Atherosclerosis Unit, Department of Internal Medicine/IMIBIC/Reina Sofia University Hospital/University of Córdoba, Córdoba, Spain; CIBER Fisiología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Elena M. Yubero-Serrano
- Lipid and Atherosclerosis Unit, Department of Internal Medicine/IMIBIC/Reina Sofia University Hospital/University of Córdoba, Córdoba, Spain; CIBER Fisiología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose M. Villalba
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario, ceiA3, Córdoba, Spain
| | - Jose Lopez-Miranda
- Lipid and Atherosclerosis Unit, Department of Internal Medicine/IMIBIC/Reina Sofia University Hospital/University of Córdoba, Córdoba, Spain; CIBER Fisiología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
44
|
Preclinical Study of the Pharmacokinetics of a New Intravenous Dosage Form of Ubiquinol. Pharm Chem J 2018. [DOI: 10.1007/s11094-018-1721-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
45
|
Fernández-Del-Río L, Nag A, Gutiérrez Casado E, Ariza J, Awad AM, Joseph AI, Kwon O, Verdin E, de Cabo R, Schneider C, Torres JZ, Burón MI, Clarke CF, Villalba JM. Kaempferol increases levels of coenzyme Q in kidney cells and serves as a biosynthetic ring precursor. Free Radic Biol Med 2017; 110:176-187. [PMID: 28603085 PMCID: PMC5539908 DOI: 10.1016/j.freeradbiomed.2017.06.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/29/2017] [Accepted: 06/06/2017] [Indexed: 12/27/2022]
Abstract
Coenzyme Q (Q) is a lipid-soluble antioxidant essential in cellular physiology. Patients with Q deficiencies, with few exceptions, seldom respond to treatment. Current therapies rely on dietary supplementation with Q10, but due to its highly lipophilic nature, Q10 is difficult to absorb by tissues and cells. Plant polyphenols, present in the human diet, are redox active and modulate numerous cellular pathways. In the present study, we tested whether treatment with polyphenols affected the content or biosynthesis of Q. Mouse kidney proximal tubule epithelial (Tkpts) cells and human embryonic kidney cells 293 (HEK 293) were treated with several types of polyphenols, and kaempferol produced the largest increase in Q levels. Experiments with stable isotope 13C-labeled kaempferol demonstrated a previously unrecognized role of kaempferol as an aromatic ring precursor in Q biosynthesis. Investigations of the structure-function relationship of related flavonols showed the importance of two hydroxyl groups, located at C3 of the C ring and C4' of the B ring, both present in kaempferol, as important determinants of kaempferol as a Q biosynthetic precursor. Concurrently, through a mechanism not related to the enhancement of Q biosynthesis, kaempferol also augmented mitochondrial localization of Sirt3. The role of kaempferol as a precursor that increases Q levels, combined with its ability to upregulate Sirt3, identify kaempferol as a potential candidate in the design of interventions aimed on increasing endogenous Q biosynthesis, particularly in kidney.
Collapse
Affiliation(s)
- Lucía Fernández-Del-Río
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario, ceiA3, Spain
| | - Anish Nag
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Elena Gutiérrez Casado
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario, ceiA3, Spain
| | - Julia Ariza
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario, ceiA3, Spain
| | - Agape M Awad
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Akil I Joseph
- Department of Pharmacology, and the Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, USA
| | - Ohyun Kwon
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Claus Schneider
- Department of Pharmacology, and the Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN, USA
| | - Jorge Z Torres
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - María I Burón
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario, ceiA3, Spain
| | - Catherine F Clarke
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - José M Villalba
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario, ceiA3, Spain
| |
Collapse
|
46
|
Pérez-Sánchez C, Aguirre MÁ, Ruiz-Limón P, Ábalos-Aguilera MC, Jiménez-Gómez Y, Arias-de la Rosa I, Rodriguez-Ariza A, Fernández-Del Río L, González-Reyes JA, Segui P, Collantes-Estévez E, Barbarroja N, Velasco F, Sciascia S, Cecchi I, Cuadrado MJ, Villalba JM, López-Pedrera C. Ubiquinol Effects on Antiphospholipid Syndrome Prothrombotic Profile: A Randomized, Placebo-Controlled Trial. Arterioscler Thromb Vasc Biol 2017; 37:1923-1932. [PMID: 28684614 DOI: 10.1161/atvbaha.117.309225] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 06/26/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Antiphospholipid syndrome (APS) leukocytes exhibit an oxidative perturbation, directly linked to alterations in mitochondrial dynamics and metabolism. This disturbance is related to the patients' prothrombotic status and can be prevented by in vitro treatment with coenzyme Q10. Our aim was to investigate short-term effects of in vivo ubiquinol (reduced coenzyme Q10 [Qred]) supplementation on markers related to inflammation and thrombosis in APS through a prospective, randomized, crossover, placebo-controlled trial. APPROACH AND RESULTS Thirty-six patients with APS were randomized to receive Qred (200 mg/d) or placebo for 1 month. Thirty-three patients with APS completed the intervention, which increased plasma coenzyme Q10. Qred improved endothelial function and decreased monocyte expression of prothrombotic and proinflammatory mediators, inhibited phosphorylation of thrombosis-related protein kinases, and decreased peroxides and percentage of monocytes with depolarized mitochondria; mitochondrial size was increased, and mitochondrial biogenesis-related genes were upregulated. Qred ameliorated extruded neutrophil extracellular traps in neutrophils and downregulated peroxides, intracellular elastase, and myeloperoxidase. Nanostring microRNA profiling revealed 20 microRNAs reduced in APS monocytes, and 16 of them, with a preponderance of cardiovascular disease-related target mRNAs, were upregulated. Monocytes gene profiling showed differential expression of 29 atherosclerosis-related genes, 23 of them changed by Qred. Interaction networks of genes and microRNAs were identified. Correlation studies demonstrated co-ordinated effects of Qred on thrombosis and endothelial function-associated molecules. CONCLUSIONS Our results highlight the potential of Qred to modulate the overexpression of inflammatory and thrombotic risk markers in APS. Because of the absence of clinically significant side effects and its potential therapeutic benefits, Qred might act as safe adjunct to standard therapies in APS. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT02218476.
Collapse
Affiliation(s)
- Carlos Pérez-Sánchez
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - María Ángeles Aguirre
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Patricia Ruiz-Limón
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - María Carmen Ábalos-Aguilera
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Yolanda Jiménez-Gómez
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Iván Arias-de la Rosa
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Antonio Rodriguez-Ariza
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Lucía Fernández-Del Río
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - José Antonio González-Reyes
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Pedro Segui
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Eduardo Collantes-Estévez
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Nuria Barbarroja
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Francisco Velasco
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Savino Sciascia
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Irene Cecchi
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - María José Cuadrado
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - José Manuel Villalba
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.)
| | - Chary López-Pedrera
- From the Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain (C.P.-S., M.Á.A., P.R.-L., M.C.A.-A., Y.J.-G., I.A.-d.l.R., A.R.A., P.S., E.C.-E., N.B., F.V., C.L.-P.); Unidad de Gestión Clínica Reumatología (M.Á.A., E.C.-E., C.L.-P.), Unidad de Gestión Clínica Radiología (P.S.), and Unidad de Gestión Clínica Hematología (F.V.), Hospital Universitario Reina Sofía, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3 (L.F.-d.R., J.A.G.-R., J.M.V.) and Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología) (E.C.-E.), Universidad de Córdoba, Córdoba, Spain; Lupus Research Unit, Hospital St Thomas, London, United Kingdom (M.J.C.); and Center of Research of Immunopathology and Rare Diseases-Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, University of Turin, Italy (S.S., I.C.).
| |
Collapse
|
47
|
Lone Y, Bhide M, Koiri RK. Amelioratory effect of coenzyme Q10 on potential human carcinogen Microcystin-LR induced toxicity in mice. Food Chem Toxicol 2017; 102:176-185. [PMID: 28219701 DOI: 10.1016/j.fct.2017.02.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 12/28/2016] [Accepted: 02/14/2017] [Indexed: 10/20/2022]
Abstract
Microcystins are a group of cyclic heptapeptide toxins produced by cyanobacteria. More than 100 microcystin analogues have been detected, among which microcystin-LR is the most abundant and toxic variant. Present study was designed to reveal whether potential human carcinogen microcystin-LR could imbalance the glycolytic-oxidative-nitrosative status of heart, kidney and spleen of mice and also to explore the amelioratory effect of coenzyme Q10 on microcystin-LR induced toxicity. Microcystin-LR was administered at a dose of 10 μg/kg bw/day, ip for 14 days in male mice. In microcystin-LR treated mice as compared to control, significant increase in the level of lipid peroxidation, hydrogen peroxide, lactate dehydrogenase, nitric oxide with a concomitant decrease in the level of glutathione was observed, suggesting microcystin-LR induced toxicity via induction of oxidative-nitrosative-glycolytic pathway. Although several studies have evaluated numerous antioxidants but still there is no effective chemoprotectant against microcystin-LR induced toxicity. When microcystin-LR treated mice were co-administered coenzyme Q10 (10 mg/kg bw/day, im) for 14 days, it was observed that coenzyme Q10 ameliorates microcystin-LR induced toxicity via modulation of glycolytic-oxidative-nitrosative stress pathway. Thus, the results suggest that coenzyme Q10 has a potential to be developed as preventive agent against microcystin-LR induced toxicity.
Collapse
Affiliation(s)
- Yaqoob Lone
- Department of Zoology, Dr. Harisingh Gour Central University, Sagar, Madhya Pradesh, 470003, India
| | - Mangla Bhide
- Department of Zoology, Dr. Harisingh Gour Central University, Sagar, Madhya Pradesh, 470003, India
| | - Raj Kumar Koiri
- Department of Zoology, Dr. Harisingh Gour Central University, Sagar, Madhya Pradesh, 470003, India.
| |
Collapse
|
48
|
Varela-López A, Giampieri F, Battino M, Quiles JL. Coenzyme Q and Its Role in the Dietary Therapy against Aging. Molecules 2016; 21:373. [PMID: 26999099 PMCID: PMC6273282 DOI: 10.3390/molecules21030373] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/12/2022] Open
Abstract
Coenzyme Q (CoQ) is a naturally occurring molecule located in the hydrophobic domain of the phospholipid bilayer of all biological membranes. Shortly after being discovered, it was recognized as an essential electron transport chain component in mitochondria where it is particularly abundant. Since then, more additional roles in cell physiology have been reported, including antioxidant, signaling, death prevention, and others. It is known that all cells are able to synthesize functionally sufficient amounts of CoQ under normal physiological conditions. However, CoQ is a molecule found in different dietary sources, which can be taken up and incorporated into biological membranes. It is known that mitochondria have a close relationship with the aging process. Additionally, delaying the aging process through diet has aroused the interest of scientists for many years. These observations have stimulated investigation of the anti-aging potential of CoQ and its possible use in dietary therapies to alleviate the effects of aging. In this context, the present review focus on the current knowledge and evidence the roles of CoQ cells, its relationship with aging, and possible implications of dietary CoQ in relation to aging, lifespan or age-related diseases.
Collapse
Affiliation(s)
- Alfonso Varela-López
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center (CIBM), University of Granada, Avda. del Conocimiento s.n., Armilla, Granada 18100, Spain.
| | - Francesca Giampieri
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche (DISCO), Facoltà di Medicina, Università Politecnica delle Marche, Ancona 60131, Italy.
| | - Maurizio Battino
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche (DISCO), Facoltà di Medicina, Università Politecnica delle Marche, Ancona 60131, Italy.
- Centre for Nutrition & Health, Universidad Europea del Atlantico (UEA), Santander 39011, Spain.
| | - 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., Armilla, Granada 18100, Spain.
| |
Collapse
|
49
|
A Therapeutic Insight of Niacin and Coenzyme Q10 Against Diabetic Encephalopathy in Rats. Mol Neurobiol 2016; 54:1601-1611. [DOI: 10.1007/s12035-016-9765-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 02/01/2016] [Indexed: 11/25/2022]
|
50
|
Li H, Chen F. Preparation and quality evaluation of coenzyme Q10 long-circulating liposomes. Saudi J Biol Sci 2015; 24:797-802. [PMID: 28490948 PMCID: PMC5415142 DOI: 10.1016/j.sjbs.2015.10.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 10/27/2015] [Accepted: 10/29/2015] [Indexed: 12/21/2022] Open
Abstract
The aim of this work was to prepare coenzyme Q10 (CoQ10) long-circulating liposomes, and establish the quality standard to determine the content and entrapment efficiency. CoQ10 long-circulating liposomes were prepared by the film dispersion method, HPLC assay for the determination of CoQ10 was developed. Free drugs and liposomes were separated using the protamine aggregation method and entrapment efficiency was determined. The liposomes were homogeneous and the mean diameter was 166.0 nm, Zeta potential was -22.2 mV. The content and entrapment efficiency of CoQ10 were 98.2% and 93.2% for three batches of liposomes, respectively. The lyophilized form of liposomes prepared by freeze-drying showed stable quality characteristics during storage. The formulation and preparative method can be used to prepare CoQ10 long-circulating liposomes with high entrapment efficiency and high quality, the determination method of drug content and entrapment efficiency were effective and rapid and can be used for quality evaluation of liposomes.
Collapse
Affiliation(s)
- Huibin Li
- Department of Burn and Plastic Surgery, People’s Hospital of Linyi, Linyi, Shandong, PR China
- Corresponding author.
| | - Fang Chen
- Department of Orthopedics, People’s Hospital of Linyi, Linyi, Shandong, PR China
| |
Collapse
|